WO2009139939A2 - Nanoparticules hybrides comme agents thérapeutiques anticancéreux et agents doubles thérapeutiques/de contraste d’imagerie - Google Patents

Nanoparticules hybrides comme agents thérapeutiques anticancéreux et agents doubles thérapeutiques/de contraste d’imagerie Download PDF

Info

Publication number
WO2009139939A2
WO2009139939A2 PCT/US2009/034867 US2009034867W WO2009139939A2 WO 2009139939 A2 WO2009139939 A2 WO 2009139939A2 US 2009034867 W US2009034867 W US 2009034867W WO 2009139939 A2 WO2009139939 A2 WO 2009139939A2
Authority
WO
WIPO (PCT)
Prior art keywords
nanoparticle
group
platinum
metal atom
cancer
Prior art date
Application number
PCT/US2009/034867
Other languages
English (en)
Other versions
WO2009139939A3 (fr
Inventor
Wenbin Lin
William J. Rieter
Original Assignee
The University Of North Carolina At Chapel Hill
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The University Of North Carolina At Chapel Hill filed Critical The University Of North Carolina At Chapel Hill
Priority to US12/918,748 priority Critical patent/US20110135571A1/en
Publication of WO2009139939A2 publication Critical patent/WO2009139939A2/fr
Publication of WO2009139939A3 publication Critical patent/WO2009139939A3/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6905Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion
    • A61K47/6907Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a microemulsion, nanoemulsion or micelle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • A61K47/6931Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer
    • A61K47/6933Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer the polymer being obtained by reactions only involving carbon to carbon, e.g. poly(meth)acrylate, polystyrene, polyvinylpyrrolidone or polyvinylalcohol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/04X-ray contrast preparations
    • A61K49/0409Physical forms of mixtures of two different X-ray contrast-enhancing agents, containing at least one X-ray contrast-enhancing agent which is not a halogenated organic compound
    • A61K49/0414Particles, beads, capsules or spheres
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/18Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes
    • A61K49/1806Suspensions, emulsions, colloids, dispersions
    • A61K49/1809Micelles, e.g. phospholipidic or polymeric micelles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/18Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes
    • A61K49/1818Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles
    • A61K49/1821Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles
    • A61K49/1824Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T428/00Stock material or miscellaneous articles
    • Y10T428/29Coated or structually defined flake, particle, cell, strand, strand portion, rod, filament, macroscopic fiber or mass thereof
    • Y10T428/2982Particulate matter [e.g., sphere, flake, etc.]

Definitions

  • the presently disclosed subject matter provides hybrid nanomaterials comprising coordination polymers prepared from metal complexes for use as anticancer agents and as combination anticancer and imaging agents.
  • CNS central nervous system
  • CTAB cetyltrimethy lammonium bromide
  • DCP direct current plasma
  • DLS dynamic light scattering
  • ICP-MS inductively coupled plasma-mass spectrometry
  • NCP nanoscale coordination polymer
  • PEG poly(ethylene glycol)
  • PET positron emission tomography
  • PVP polyvinylpyrrolidone
  • RES reticuloendothelial system
  • RGD arginine-glycine-aspartic acid
  • TEOS tetraethyl orthosilicate
  • UV ultraviolet
  • W water to surfactant molar ratio
  • wt% weight percentage
  • XRT X-ray radiation
  • a variety of anticancer drugs are available for treating different types of cancers in the clinic.
  • the therapeutic efficacy of these drugs is often limited by the inability to selectively deliver the drugs to tumors.
  • Most of the currently available anticancer drugs are highly cytotoxic, and can kill normal cells along with cancerous cells. Thus, when high doses of drugs are used, there can be severe side effects.
  • most of the currently used anticancer drugs have a rather limited therapeutic index.
  • Such a limit on dosage prevents the complete eradication of cancer cells in a patient, and can lead to recurrence of the cancer in many patients.
  • the limit in dosage can also predispose the recurring cancer to drug resistance, thus worsening the prognosis for the patient.
  • the presently disclosed subject matter provides a nanoparticle comprising a coordination polymer comprising a plurality of platinum metal complexes.
  • the plurality of platinum metal complexes include but are not limited to a plurality of platinum (II) metal complexes, a plurality of platinum (IV) metal complexes, or a combination thereof.
  • one or more of the platinum metal complexes comprises: a platinum metal atom; at least one nonbridging ligand, wherein the at least one nonbridging ligand is bonded to the platinum metal atom through at least one coordination bond; and at least one bridging ligand, wherein the at least one bridging ligand is bonded to the platinum metal atom through at least one coordination bond and comprises at least one linking moiety, wherein each of the at least one linking moiety is bonded to an additional metal atom via a coordination bond.
  • each of the at least one linking moiety is independently selected from the group consisting of a carboxylate, a carboxylic acid, an amine, a hydroxyl, a thiol, a carbamate, an ester, a phosphate, a phosphonate, a carbonate, and an amide.
  • each of the at least one bridging ligand is independently selected from the group consisting of a polymeric bridging ligand and a nonpolymeric bridging ligand. In some embodiments, each of the at least one bridging ligand is a nonpolymeric bridging ligand. In some embodiments, the bridging ligand comprises at least two carboxylate groups.
  • At least one platinum metal complex comprises two bridging ligands, wherein each of the two bridging ligands is bonded to the platinum metal atom through one coordination bond and comprises at least one linking moiety.
  • each of the two bridging ligands is independently selected from the group consisting of 1 ,4-benzene dicarboxylate;
  • At least one platinum metal complex comprises one bridging ligand, wherein the one bridging ligand is bonded to the platinum metal atom through two coordination bonds and comprises at least two linking moieties.
  • the one bridging ligand is a bipyridine dicarboxylate.
  • the bipyridine dicarboxylate is selected from 2,2'-bipyridine-5,5'-dicarboxylate and 2,2'-bipyridine-4,4'-dicarboxylate.
  • one of the at least one bridging ligand is a nonplatinum anticancer drug.
  • the nonplatinum anticancer drug is selected from the group consisting of methotrexate, folic acid, leucovorin, vinblastine, vincristine, melphalan, imatinib, pemetrexed, vindesine, anastrozole, doxorubicin, cytarabine, azathioprine, letrozole and carboxylates thereof.
  • one of the at least one bridging ligand is a polymeric bridging ligand.
  • the polymeric bridging ligand comprises one of the group consisting of poly(silsesquioxane), poly(siloxane), poly(acrylate) and poly(acrylamide).
  • the additional metal atom is a platinum metal atom of a second platinum metal complex.
  • the additional metal atom is a nonplatinum metal atom selected from the group consisting of a transition metal atom, a lanthanide metal atom, and an actinide metal atom.
  • the additional metal atom is selected from the group consisting of Tb 3+ and Zn 2+ .
  • each of the at least one nonbridging ligands is independently selected from the group consisting of NH 3 , a primary amine, a secondary amine, a diamine, an aromatic amine, a halide, and hydroxide.
  • the diamine is a cyclohexanediamine.
  • each of the at least one nonbridging ligands is independently selected from the group consisting of NH 3 and chloride.
  • each of the plurality of platinum metal complexes is independently selected from the group consisting of:
  • the nanoparticle has a diameter ranging between about 20 nm and about 250 nm. In some embodiments, the nanoparticle has a diameter ranging between about 40 nm and about 70 nm.
  • the nanoparticle further comprises one or more of the group consisting of a photosensitizer, a radiosensitizer, a radionuclide, a passivating agent, an imaging agent, and a targeting agent.
  • the imaging agent can be selected from the group consisting of an optical imaging agent, a magnetic resonance imaging (MRI) agent, a positron emission tomography
  • the optical imaging agent is a luminescent agent.
  • the targeting agent is selected from the group consisting of a small molecule, a peptide, and a protein. In some embodiments, the targeting agent binds to a receptor or ligand present on a cancer cell. In some embodiments, the targeting agent comprises cyclic(RGDfk).
  • an outer surface of the nanoparticle is chemically modified with one or more of a passivating agent, a targeting agent, and an imaging agent.
  • the passivating agent comprises poly(ethylene glycol).
  • the nanoparticle comprises a core and an outer layer, the core comprising a coordination polymer comprising a plurality of platinum metal complexes, and the outer layer surrounding the core and comprising one or more of a metal oxide, a lipid bilayer, an organic polymer, a silica-based polymer, and combinations thereof.
  • the organic polymer is polyvinylpyrolidone (PVP).
  • the outer layer is polyvinylpyrolidone (PVP), Si ⁇ 2 , or a combination thereof.
  • the outer layer has a thickness ranging between about 1 nm and about 10 nm.
  • the presently disclosed subject matter provides a pharmaceutical composition comprising a nanoparticle comprising a coordination polymer comprising a plurality of platinum metal complexes and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition is pharmaceutically acceptable in humans.
  • the pharmaceutical composition comprises one of a liposome and a microemulsion.
  • the presently disclosed subject matter provides a method of inhibiting proliferation of a cancer cell, the method comprising contacting the cancer cell with a nanoparticle, wherein the nanoparticle comprises a coordination polymer comprising a plurality of platinum metal complexes.
  • the cancer cell is selected from a skin cancer cell, a connective tissue cancer cell, an esophageal cancer cell, a head and neck cancer cell, a breast cancer cell, a lung cancer cell, a stomach cancer cell, a pancreatic cancer cell, an ovarian cancer cell, a cervical cancer cell, a uterine cancer cell, an anogenital cancer cell, a kidney cancer cell, a bladder cancer cell, a colon cancer cell, a prostate cancer cell, a retinal cancer cell, a central nervous system cancer cell, and a lymphoid cancer cell.
  • the presently disclosed subject matter provides a method of treating cancer in a subject in need of treatment thereof, the method comprising administering to the subject a nanoparticle comprising a coordination polymer, wherein the coordination polymer comprises a plurality of platinum metal complexes.
  • the cancer is selected from a skin cancer, a connective tissue cancer, an esophageal cancer, a head and neck cancer, a breast cancer, a lung cancer, a stomach cancer, a pancreatic cancer, an ovarian cancer, a cervical cancer, a uterine cancer, an anogenital cancer, a kidney cancer, a bladder cancer, a colon cancer, a prostate cancer, a retinal cancer, a central nervous system cancer, and a lymphoid cancer.
  • the subject is a mammal.
  • the method further comprises imaging delivery of the nanoparticle in one or more tissue or organ in the subject following administration of the nanoparticle. In some embodiments, the method further comprises administering to the subject an external stimulus selected from the group consisting of laser light and X-ray radiation.
  • the presently disclosed subject matter provides a method of synthesizing a nanoparticle comprising a coordination polymer comprising a plurality of platinum metal complexes, wherein the method comprises precipitation or use of a microemulsion system.
  • the method comprises providing a solution comprising a first solvent, at least one bridging ligand precursor, and a plurality of platinum diaqua complexes selected from the group consisting of platinum
  • the method further comprises adjusting the pH of the solution prior to adding the second solvent.
  • the first solvent comprises water, dimethyl sulfoxide (DMSO), or a combination thereof.
  • the at least one bridging ligand precursor is selected from the group consisting of a benzene dicarboxylic acid, a benzene dicarboxylate, a carboxylate-substituted styrene, a carboxylate-substituted silyl ether, a bipyridine dicarboxylic acid, a bipyridine dicarboxylate, a dicarboxylic anhydride, a diacyl dichloride, and a nonplatinum anticancer drug.
  • the second solvent is selected from the group consisting of acetone, an alcohol, ether, and acetonitrile.
  • the solution further comprises an additional component, wherein the additional component is selected from the group consisting of a radionuclide, an imaging agent, a photosensitizer, and a radiosensitizer, and adding the second solvent co-precipitates the additional component, thereby incorporating the additional component into the nanoparticle.
  • the method comprises providing a first mixture comprising a microemulsion system comprising water, an organic solvent, a surfactant, and a co-surfactant; adding to the first mixture an aqueous solution comprising a platinum metal complex, thereby forming a second mixture, wherein the platinum metal complex comprises a platinum metal atom, one or more nonbridging ligands, and at least one ligand bound to the platinum metal atom by at least one coordination bond and comprising at least one prelinking moiety, wherein the at least one prelinking moiety is a group that can form a coordination bond with an additional metal atom; stirring the second mixture until the second mixture is visably clear; providing a third mixture comprising a microemulsion system comprising water, an organic solvent, a surfactant, and a co-surfactant; adding to the third mixture an aqueous solution comprising a nonplatinum metal compound, thereby forming a fourth mixture; stirring the fourth mixture until the fourth mixture is visably clear; adding the
  • the method comprises providing a microemulsion system comprising water; an organic solvent; a surfactant; a co-surfactant; a polymerizable monomer; and a platinum metal complex, wherein the platinum metal complex comprises a platinum metal atom, one or more nonbridging ligands, and at least one ligand bonded to the platinum metal atom by at least one coordination bond and comprising at least one prelinking moiety, where the at least one prelinking moiety is a moiety that can react with the polymerizable monomer.
  • the prelinking moiety is selected from the group consisting of an alkyl halide, an acyl halide, a silyl ether, an alkene, an alkyne, a carboxylic acid, an amine, an ester, an anhydride, and an isocyanate.
  • the polymerizable monomer is selected from the group consisting of a silyl ether, acrylic acid, and acrylamide.
  • the method further comprises isolating the nanoparticle via centrifugation. In some embodiments, the method further comprises coating the nanoparticle with one or more of the group consisting of a metal oxide, a lipid bilayer, an organic polymer, a silica-based polymer, and combinations thereof. In some embodiments, the method further comprises grafting onto a surface of the nanoparticle one or more of a photosensitizer, a radiosensitizer, a radionuclide, an imaging agent, a passivating agent, and a targeting agent.
  • the presently disclosed subject matter provides a coordination polymer comprising a plurality of platinum metal complexes wherein the platinum metal complexes are linked via bridging ligands, wherein each bridging ligand is independently selected from the group consisting of a nonpolymeric bridging ligand and a polymeric bridging ligand.
  • the presently disclosed subject matter provides a coordination polymer comprising a plurality of nonplatinum metal complexes wherein the nonplatinum metal complexes are linked via bridging ligands, wherein one or more of the bridging ligands are a nonplatinum anticancer drug. It is an object of the presently disclosed subject matter to provide anticancer therapeutic agents and dual anticancer/imaging contrast agents. An object of the presently disclosed subject matter having been stated hereinabove, and which is achieved in whole or in part by the presently disclosed subject matter, other objects will become evident as the description proceeds when taken in connection with the accompanying drawings as best described hereinbelow.
  • Figure 1 is a drawing showing the structures of platinum (II) anticancer prodrugs and their active diaqua complexes.
  • Figure 2 is a schematic drawing showing the synthesis of a platinum (II) metal complex-based coordination polymer.
  • Figure 3 is a schematic drawing showing the synthesis of a platinum (IV) metal complex-based coordination polymer.
  • Figure 4 is a schematic drawing showing the synthesis of a coordination polymer comprising both platinum (II) and platinum (IV) metal complexes.
  • Figure 5 is a series of drawings showing the structures of exemplary small molecule, organic anticancer drugs that can be used as bridging ligands in coordination polymer nanoparticles.
  • Figure 6 is a series of three schematic diagrams showing the synthesis of nanoparticles comprising coordination polymers comprising platinum metal complexes having polymeric bridging ligands.
  • Figure 7 are two drawings showing the structures of exemplary photo- and radiosensitizers that can be incorporated into coordination polymer nanoparticles.
  • Figure 8 is a schematic diagram showing the synthesis of a silica layer stabilized, C(RGDf K)-targeted, bimetallic coordination polymer nanoparticle.
  • Figure 9 are scanning electron micrograph (SEM) images of cis- Pt(NH 3 ) 2 (benzene dicarboxylate) nanoclusters synthesized via the rapid addition of acetone to a precursor (aqueous) solution. Scale markings in the left-hand image are every 5 microns. Scale markings in the right-hand image are every 1 micron.
  • Figure 10 are scanning electron micrograph (SEM) images of cis-
  • Pt(NH 3 ) 2 (benzene dicarboxylate) nanoclusters synthesized via the rapid addition of an acetone/ethanol mixture (1 :1 v/v) to a precursor (aqueous) solution.
  • Scale markings in the left-hand image are every 10 microns.
  • Scale markings in the right-hand image are every 3 microns.
  • Figure 11 are scanning electron micrograph (SEM) images of Tb x [c,c,f- Pt(NH 3 )2(CI)2( ⁇ 2CCH2CH 2 C ⁇ 2)2]y nanoparticles synthesized via cationic microemulsions with [water]/[surfactant] ratios of 15 (left image) and 20 (right image). Scale markings in both images are every 5 nm.
  • Figure 12 are transmission electron micrograph (TEM) images of Tb x [c,c,f-Pt(NH 3 ) 2 (CI) 2 (O 2 CCH 2 CH 2 CO 2 ) 2 ]y nanoparticles (NCP-1) synthesized via the rapid addition of an methanol to a precursor (aqueous) solution.
  • the scale marking in the left-hand corner of the left-hand image represents 0.2 microns.
  • the scale marking in the left-hand corner of the right-hand image represents 50 nm.
  • Figure 13 are scanning electron micrograph (SEM) images of Tb x [C, c,t- Pt(NH 3 )2(CI)2( ⁇ 2CCH2CH2CO 2 )2]y nanoparticles synthesized via the rapid addition of an ethanol/methanol mixture (1 :1 v/v) to a precursor (aqueous) solution.
  • Figure 14 are scanning electron micrograph (SEM) images of Zn x [C, c,t- Pt(NH 3 )2(CI)2( ⁇ 2CCH2CH2CO 2 )2]y nanoparticles synthesized via the rapid addition of methanol to a precursor (aqueous) solution.
  • Figure 15 are scanning electron micrograph (SEM) images of poly(vinylpyrrolidone) (PVP) coated Tb x [cc>Pt(NH 3 ) 2 (CI) 2 (O 2 CCH 2 CH 2 CO 2 ) 2 ] y nanoparticles (NCP-1/PVP). The scale marking in the left-hand corner of the left-hand image represents 0.2 microns.
  • FIG. 16A is a transmission electron micrograph (TEM) image of silica- coated Tb x [cc ⁇ Pt(NH 3 ) 2 (CI) 2 (O 2 CCH 2 CH 2 CO 2 ) 2 ] y nanoparticles (i.e., NCP-1 ') isolated after 2 hours of exposure to a silica-coating solution.
  • the scale bar in the left-hand corner of the image represents 50 nm.
  • Figure 16B is a transmission electron micrograph (TEM) image of silica- coated Tb x [C, c ; f-Pt(NH 3 ) 2 (CI) 2 ( ⁇ 2 CCH 2 CH 2 C ⁇ 2 ) 2 ]ynanoparticles (i.e., NCP-T) isolated after 3 hours of exposure to a silica-coating solution.
  • the scale bar in the left-hand corner of the image represents 50 nm.
  • Figure 16C is a transmission electron micrograph (TEM) image of silica- coated Tb x [C 1 c, f-Pt(NH 3 )2(CI)2( ⁇ 2CCH 2 CH 2 C ⁇ 2 ) 2 ]y nanoparticles (i.e., NCP-f) isolated after 4 hours of exposure to a silica-coating solution.
  • the scale bar in the left-hand corner of the image represents 50 nm.
  • Figure 16D is a transmission electron micrograph (TEM) image of silica- coated Tb x [C, c>Pt(NH 3 ) 2 (CI)2( ⁇ 2CCH 2 CH 2 CO 2 ) 2 ]y nanoparticles (i.e., NCP-T) isolated after 7 hours of exposure to a silica-coating solution.
  • the scale bar in the left-hand corner of the image represents 50 nm.
  • Figure 17 is a graph showing the dynamic light scattering (DLS) curves for Tb x [c,c>Pt(NH 3 ) 2 (CI) 2 (O 2 CCH 2 CH 2 CO 2 ) 2 ] y nanoparticles (NCP-1, lighter shaded circles), polyvinylpyrrolidone(PVP)-coated NCP-1 (NCP-1/PVP, more darkly shaded circles), silica-coated-NCP-1 having a 2 nm thick silica coating (NCP-T-a, triangles), and silica-coated-NCP-1 having a 7 nm thick silica coating (NCP-T-b, darkly shaded squares).
  • DLS dynamic light scattering
  • Figure 18 is a graph showing the thermogravimetric analysis (TGA) curves for the c,c,f-Pt(NH 3 )2(CI)2( ⁇ 2CCH2CH 2 C ⁇ 2)2 molecular complex
  • DSCP Tb x [c,c ) f-Pt(NH3)2(CI)2( ⁇ 2CCH 2 CH2C ⁇ 2)2]y nanoparticles
  • NCP-1 silica-coated Tb x [C, c,f-Pt(NH3)2(CI)2(O 2 CCH 2 CH 2 CO2)2]y nanoparticles
  • Figure 19 is a graph showing the release profiles for Tb x [c,c,t-
  • NCP-1 having a 7 nm thick silica coating (NCP-T-b, triangles) plotted as the %
  • Figure 20 is a graph showing the in vitro cytotoxicity assay curves for human colon cancer cells (HT-29 cells) obtained by plotting the % cell viability against the Pt concentration for cisplatin (darker diamonds), the c,c,t-
  • Tb x [C, c,f-Pt(NH3) 2 (CI) 2 ( ⁇ 2CCH2CH 2 C ⁇ 2) 2 ]y nanoparticles (NCP-1, triangles), RGD-targeted, silica-coated (2 nm thickness) Tb x [c,c,t- Pt(NH3)2(CI)2(O 2 CCH 2 CH 2 CO2)2]y nanoparticles (NCP-1'-a-c(RGDfK, circles ), and RGD-targeted, silica-coated (7 nm thickness) Tb x [C, c,t- Pt(NH3)2(CI)2( ⁇ 2CCH 2 CH2C ⁇ 2)2]y nanoparticles (NCP-1'-b-c(RGDfK, lighter shaded diamonds).
  • Figure 21A is a graph showing the in vitro cytotoxicity assay curves for human breast cancer cells (MCF-7 cells) obtained by plotting the % cell viability against the Pt concentration for cisplatin (circles) and for the c,c,t- Pt(NH 3 )2(CI)2(O 2 CCH 2 CH2CO2)2 molecular complex (DSCP, diamonds).
  • Figure 21 B is a graph showing the in vitro cytotoxicity assay curves for human breast cancer cells (MCF-7 cells) obtained by plotting the % cell viability against the Pt concentration for cisplatin (circles) -and silica-coated (7 nm thickness) Tb x [c,c>Pt(NH 3 ) 2 (CI) 2 (O 2 CCH 2 CH 2 CO 2 ) 2 ] y nanoparticles (NCP-1'-b, diamonds).
  • the term "about”, when referring to a value or to an amount of size (i.e., diameter), weight, concentration, or percentage is meant to encompass variations of, in some embodiments ⁇ 20%, in some embodiments ⁇ 10%, in some embodiments ⁇ 5%, in some embodiments ⁇ 1%, and in some embodiments ⁇ 0.1% from the specified amount, as such variations are appropriate to perform the disclosed methods.
  • nanoscale refers to a structure having at least one region with a dimension (e.g., length, width, diameter, etc.) of less than about 1 ,000 nm.
  • the dimension is smaller (e.g., less than about 500 nm, less than about 250 nm, less than about 200 nm, less than about 150 nm, less than about 125 nm, less than about 100 nm, less than about 80 nm, less than about 70 nm, less than about 60 nm, less than about 50 nm, less than about 40 nm, less than about 30 nm or even less than about 20 nm). In some embodiments, the dimension is less than about 10 nm.
  • the nanoparticle is approximately spherical.
  • the characteristic dimension can correspond to the diameter of the sphere.
  • the nanoparticle or other nanoscale material can be disc-shaped, oblong, polyhedral, rod-shaped, cubic, or irregularly-shaped.
  • a nanoscale material can also be irregularly shaped or comprise clusters of spheres, rods, discs, or cubes.
  • the nanoparticle can comprise an interior region (i.e., the space between the outer dimensions of the particle) and an outer surface (i.e., the surface that defines the outer dimensions of the particle).
  • the particle can comprise one or more layers.
  • a spherical nanoparticle can comprise one or more concentric layers, each successive layer being dispersed over the outer surface of the smaller layer closer to the center of the particle.
  • the particle can be solid or porous or can contain a hollow interior region.
  • the nanoparticle can comprise two layers, an inner core and an outer layer or shell dispersed over the core.
  • polymer and “polymeric” refer to chemical structures that have repeating units (i.e., multiple copies of a given chemical substructure). As used herein, polymers can refer to groups having more than 10 repeating units and/or to groups wherein the repeating unit is other than methylene. Polymers can be formed from polymerizable monomers.
  • a polymerizable monomer is a molecule that comprises one or more reactive moieties ⁇ e.g., siloxy ethers, hydroxyls, amines, vinylic groups (i.e., carbon-carbon double bonds), halides (i.e., Cl, Br, F, and I), esters, activated esters, and the like ⁇ that can react to form bonds with other molecules.
  • each polymerizable monomer molecule can bond to two or more other molecules. In some cases, a polymerizable monomer will bond to only one other molecule, forming a terminus of the polymeric material.
  • Exemplary polymerizable monomers include, but are not limited to, acrylamide, acrylic acid, and silyl ethers.
  • Polymers can be organic, or inorganic, or a combination thereof. As used herein, the term "inorganic" refers to a compound or composition that contains at least some atoms other than carbon, hydrogen, nitrogen, oxygen, sulfur, phosphorous, or one of the halides. Thus, for example, an inorganic polymer can contain one or more silicon atoms or one or more metal atoms in repeating units.
  • Organic polymers do not include silica or metal atoms in their repeating units.
  • Exemplary organic polymers include polyvinylpyrrolidone (PVO), polyesters, polyamides, polyethers, polydienes, and the like.
  • Some organic polymers contain biodegradable linkages, such as esters or amides, such that they can degrade overtime under biological conditions.
  • Polymers can also comprise coordination complexes.
  • the coordination complexes are repeating units in the polymer, the polymer can be referred to as a coordination polymer.
  • the terms "coordination complex” and "metal coordination complex” refer to a chemical species in which there is a coordinate bond between a metal ion and one or more ligands, wherein each ligand comprises an electron pair donor (i.e., chelating group).
  • chelating groups are generally electron pair donors, molecules or molecular ions having unshared electron pairs available for donation to a metal ion.
  • bonding or “bonded” and variations thereof can refer to either covalent or non-covalent bonding. In some cases, the term “bonding” refers to bonding via a coordinate bond.
  • coordinate bond or “coordination bond” refer to an interaction between an electron pair donor and a coordination site on a metal ion resulting in an attractive force between the electron pair donor and the metal ion.
  • coordinate bond refers to an interaction between an electron pair donor and a coordination site on a metal ion resulting in an attractive force between the electron pair donor and the metal ion.
  • the use of this term is not intended to be limiting, in so much as certain coordinate bonds also can be classified as have more or less covalent character (if not entirely covalent character) depending on the characteristics of the metal ion and the electron pair donor.
  • chelating agent refers to a molecule or molecular ion or species having an unshared electron pair available for donation to a metal ion.
  • the metal ion is coordinated by two or more electron pairs to the chelating agent.
  • “pentadentate” refer to chelating agents having two, three, four, and five electron pairs, respectively, available for simultaneous donation to a metal ion coordinated by the chelating agent.
  • the electron pairs of a chelating agent form coordinate bonds with a single metal ion.
  • the electron pairs of a chelating agent form coordinate bonds with more than one metal ion, with a variety of binding modes being possible.
  • nonbridging ligand refers to a ligand in a coordination polymer that is bonded to a single metal atom.
  • the nonbridging ligand can be monodentate or bidentate.
  • Exemplary nonbridging ligands include, but are not limited to, NH 3 , primary amines, secondary amines, diamines, aromatic amines, halides, hydroxide, thiols, and water.
  • bridging ligand refers to a ligand in a coordination polymer that is bonded to more than one metal atom (including, but not limited to, metal atoms in more than one repeating metal complex of the polymer).
  • a bridging ligand can refer to a ligand that is bonded to the platinum metal atoms of two different platinum metal complexes in a coordination polymer comprising a plurality of platinum metal complexes. In some embodiments, the bridging ligand is bonded to the platinum metal atom of one platinum metal complex in a coordination polymer comprising a plurality of platinum metal complexes and at least one other additional metal atom.
  • ligand can also refer to biological ligands, such as chemical entities (i.e., groups or whole molecules) that bind to biologically relevant receptors.
  • cancer refers to diseases caused by uncontrolled cell division and the ability of cells to metastasize, or to establish new growth in additional sites.
  • malignant refers to cancerous cells or groups of cancerous cells.
  • cancers include, but are not limited to, skin cancers (e.g., melanoma), connective tissue cancers (e.g., sarcomas), adipose cancers, breast cancers, head and neck cancers, lung cancers (e.g., mesothelioma), stomach cancers, pancreatic cancers, ovarian cancers, cervical cancers, uterine cancers, anogenital cancers (e.g., testicular cancer), kidney cancers, bladder cancers, colon cancers, prostate cancers, central nervous system (CNS) cancers, retinal cancer, blood, neuroblastomas, multiple myeloma, and lymphoid cancers (e.g., Hodgkin's and non-Hodgkin's lymophomas).
  • skin cancers e.g., melanoma
  • connective tissue cancers e.g., sarcomas
  • adipose cancers e.g., breast
  • anticancer drug and “anticancer prodrug” refer to drugs or prodrugs known to, or suspected of being able to treat a cancer (i.e., to kill cancer cells, prohibit proliferation of cancer cells, or treat a symptom related to cancer).
  • an imaging agent refers to a chemical moiety that aids in the visualization of a sample.
  • an imaging agent can be a "contrast agent”, and can refer to a moiety (a specific part of or an entire molecule, macromolecule, coordination complex, or nanoparticle) that increases the contrast of a biological tissue or structure being examined.
  • the contrast agent can increase the contrast of a structure being examined using magnetic resonance imaging (MRI), optical imaging, positron emission tomography (PET) imaging, single photon emission computed tomography (SPECT) imaging, or a combination thereof (i.e., the contrast agent can be multimodal).
  • MRI contrast agent or MRI imaging agent” refer to a moiety that effects a change in induced relaxation rates of water protons in a sample.
  • MRI contrast agents typically employ paramagnetic metal ions to effect such changes.
  • paramagnetic metal ion refers to a metal ion that is magnetized parallel or antiparallel to a magnetic field to an extent proportional to the field.
  • paramagnetic metal ions are metal ions that have unpaired electrons.
  • Paramagnetic metal ions can be selected from the group (including, but not limited to, transition and inner transition elements, including, but not limited to, scandium, titanium, vanadium, chromium, cobalt, nickel, copper, molybdenum, ruthenium, cerium, praseodymium, neodymium, promethium, samarium, europium, terbium, holmium, erbium, thulium, and ytterbium.
  • transition and inner transition elements including, but not limited to, scandium, titanium, vanadium, chromium, cobalt, nickel, copper, molybdenum, ruthenium, cerium, praseodymium, neodymium, promethium, samarium, europium, terbium, holmium, erbium, thulium, and ytterbium.
  • the paramagnetic metal ions can be selected from the group (including, but not limited to, gadolinium III (i.e., Gd +3 or Gd(III)); manganese Il (i.e., Mn +2 or Mn(II)); copper Il (i.e., Cu +2 or Cu(II)); chromium III (i.e., Cr +3 or Cr(III)); iron Il (i.e., Fe +2 or Fe(II)); iron III (i.e., Fe +3 or Fe(III)); cobalt Il (i.e., Co +2 or Co(II)); erbium Il (i.e., Er +2 or Er(II)), nickel Il (i.e., Ni +2 or Ni(II)); europium III (i.e., Eu +3 or Eu(III)); yttrium III (i.e., Yt +3 or Yt(III)); and dysprosium III (i.e.,
  • optical imaging agent or “optical contrast agent” refer to a group that can be detected based upon an ability to absorb, reflect or emit light (e.g., ultraviolet, visible, or infrared light). Optical imaging agents can be detected based on a change in amount of absorbance, reflectance, or fluorescence, or a change in the number of absorbance peaks or their wavelength maxima. Thus, optical imaging agents include those which can be detected based on fluorescence or luminescence, including organic and inorganic dyes.
  • Luminescence occurs when a molecule (or other chemical species) in an electronically excited state relaxes to a lower energy state by the emission of a photon.
  • the luminescent agent in one embodiment can be a chemiluminescent agent.
  • the excited state is generated as a result of a chemical reaction, such as lumisol and isoluminol.
  • photoluminescence such as fluorescence and phosphorescence
  • an electronically excited state is generated by the illumination of a molecule with an external light source.
  • Bioluminescence can occur as the result of action by an enzyme, such as luciferase.
  • electrochemiluminescence In electrochemiluminescence (ECL), the electronically excited state is generated upon exposure of the molecule (or a precursor molecule) to electrochemical energy in an appropriate surrounding chemical environment.
  • electrochemiluminescent agents are provided, for example, in U.S. Patent Nos. 5,147,806 and 5,641 ,623; and in U.S. Patent Application Publication No.2001/0018187; and include, but are not limited to, metal cation-liquid complexes, substituted or unsubstituted polyaromatic molecules, and mixed systems such as aryl derivatives of isobenzofurans and indoles.
  • the electrochemiluminescent chemical moiety can comprise, in a specific embodiment, a metal-containing organic compound wherein the metal is selected from the group consisting of ruthenium, osmium, rhenium, iridium, rhodium, platinum, palladium, molybdenum, technetium and tungsten.
  • ruthenium osmium, rhenium, iridium, rhodium, platinum, palladium, molybdenum, technetium and tungsten.
  • fluorophore refers to a species that can be excited by visible light or non-visible light (e.g., UV light).
  • fluorophores include, but are not limited to: quantum dots and doped quantum dots (e.g., a semiconducting CdSe quantum dot or a Mn-doped CdSe quantum dot), fluorescein, fluorescein derivatives and analogues, indocyanine green, rhodamine, triphenylmethines, polymethines, cyanines, phalocyanines, naphthocyanines, merocyanines, lanthanide complexes or cryptates, fullerenes, oxatellurazoles, LaJoIIa blue, porphyrins and porphyrin analogues and natural chromophores/fluorophores such as chlorophyll, carotenoids, flavonoids, bilins, phytochrome, phycobilins, phycoerythrin, phycocyanines, retinoic acid and analogues such as retinoins and retinates.
  • quantum dot refers to semiconductors comprising an inorganic crystalline material that is luminescent (i.e., that is capable of emitting electromagnetic radiation upon excitation).
  • the quantum dot can include an inner core of one or more first semiconductor materials that is optionally contained within an overcoating or "shell" of a second semiconductor material.
  • a semiconductor nanocrystal core surrounded by a semiconductor shell is referred to as a "core/shell” semiconductor nanocrystal.
  • the surrounding shell material can optionally have a bandgap energy that is larger than the bandgap energy of the core material and can be chosen to have an atomic spacing close to that of the core substrate.
  • Suitable semiconductor materials for quantum dots include, but are not limited to, materials comprising a first element selected from Groups 2 and 12 of the Periodic Table of the Elements and a second element selected from Group 16. Such materials include, but are not limited to ZnS, ZnSe, ZnTe, CDs, CdSe, CdTe, HgS, HgSe, HgTe, MgS, MgSe, MgTe, CaS, CaSe, CaTe, SrS, SrSe, SrTe, BaS, BaSe, BaTe, and the like. Suitable semiconductor materials also include materials comprising a first element selected from Group 13 of the Periodic Table of the Elements and a second element selected from Group 15. Such materials include, but are not limited to, GaN, GaP, GaAs,
  • Semiconductor materials further include materials comprising a Group 14 element (Ge, Si, and the like); materials such as PbS, PbSe and the like; and alloys and mixtures thereof.
  • a Group 14 element Ge, Si, and the like
  • materials such as PbS, PbSe and the like
  • alloys and mixtures thereof As used herein, all reference to the Periodic Table of the Elements and groups thereof is to the new IUPAC system for numbering element groups, as set forth in the Handbook of Chemistry and Physics, 81st Edition (CRC Press, 2000).
  • alkyl refers to Ci -2 o inclusive, linear (i.e.,
  • straight-chain branched, or cyclic, saturated or at least partially and in some cases fully unsaturated (i.e., alkenyl and alkynyl) hydrocarbon chains, including for example, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, ferf-butyl, pentyl, hexyl, octyl, ethenyl, propenyl, butenyl, pentenyl, hexenyl, octenyl, butadienyl, propynyl, butynyl, pentynyl, hexynyl, heptynyl, and allenyl groups.
  • alkenyl and alkynyl alkenyl and alkynyl hydrocarbon chains
  • Branched refers to an alkyl group in which a lower alkyl group, such as methyl, ethyl or propyl, is attached to a linear alkyl chain.
  • Lower alkyl refers to an alkyl group having 1 to about 8 carbon atoms (i.e., a Ci -8 alkyl), e.g., 1 , 2, 3, 4, 5, 6, 7, or 8 carbon atoms.
  • Higher alkyl refers to an alkyl group having about 10 to about 20 carbon atoms, e.g., 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, or 20 carbon atoms.
  • alkyl refers, in particular, to Ci -S straight- chain alkyls.
  • alkyl refers, in particular, to Ci_ 8 branched-chain alkyls.
  • Alkyl groups can optionally be substituted (a "substituted alkyl") with one or more alkyl group substituents, which can be the same or different.
  • alkyl group substituent includes but is not limited to alkyl, substituted alkyl, halo, arylamino, acyl, hydroxyl, aryloxyl, alkoxyl, alkylthio, arylthio, aralkyloxyl, aralkylthio, carboxyl, alkoxycarbonyl, oxo, and cycloalkyl.
  • alkyl chain There can be optionally inserted along the alkyl chain one or more oxygen, sulfur or substituted or unsubstituted nitrogen atoms, wherein the nitrogen substituent is hydrogen, lower alkyl (also referred to herein as "alkylaminoalkyl”), or aryl.
  • substituted alkyl includes alkyl groups, as defined herein, in which one or more atoms or functional groups of the alkyl group are replaced with another atom or functional group, including for example, alkyl, substituted alkyl, halogen, aryl, substituted aryl, alkoxyl, hydroxyl, nitro, amino, alkylamino, dialkylamino, sulfate, and mercapto.
  • aryl is used herein to refer to an aromatic substituentthat can be a single aromatic ring, or multiple aromatic rings that are fused together, linked covalently, or linked to a common group, such as, but not limited to, a methylene or ethylene moiety.
  • the common linking group also can be a carbonyl, as in benzophenone, or oxygen, as in diphenylether, or nitrogen, as in diphenylamine.
  • aryl specifically encompasses heterocyclic aromatic compounds.
  • the aromatic ring(s) can comprise phenyl, naphthyl, biphenyl, diphenylether, diphenylamine and benzophenone, among others.
  • aryl means a cyclic aromatic comprising about 5 to about 10 carbon atoms, e.g., 5, 6, 7, 8, 9, or 10 carbon atoms, and including 5- and 6-membered hydrocarbon and heterocyclic aromatic rings.
  • the aryl group can be optionally substituted (a "substituted aryl") with one or more aryl group substituents, which can be the same or different, wherein "aryl group substituent” includes alkyl, substituted alkyl, aryl, substituted aryl, aralkyl, hydroxyl, alkoxyl, aryloxyl, aralkyloxyl, carboxyl, acyl, halo, nitro, alkoxycarbonyl, aryloxycarbonyl, aralkoxycarbonyl, acyloxyl, acylamino, aroylamino, carbamoyl, alkylcarbamoyl, dialkylcarbamoyl, arylthio, alkylthio, alkylene, and -NR 1 R", wherein R' and R" can each be independently hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, and aralkyl.
  • substituted aryl includes aryl groups, as defined herein, in which one or more atoms or functional groups of the aryl group are replaced with another atom or functional group, including for example, alkyl, substituted alkyl, halogen, aryl, substituted aryl, alkoxyl, hydroxyl, nitro, amino, alkylamino, dialkylamino, sulfate, and mercapto.
  • aryl groups include, but are not limited to, cyclopentadienyl, phenyl, furan, thiophene, pyrrole, pyran, pyridine, imidazole, benzimidazole, isothiazole, isoxazole, pyrazole, pyrazine, triazine, pyrimidine, quinoline, isoquinoline, indole, carbazole, and the like.
  • Alkylene refers to a straight or branched bivalent aliphatic hydrocarbon group having from 1 to about 20 carbon atoms, e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, or 20 carbon atoms.
  • the alkylene group can be straight, branched or cyclic.
  • the alkylene group also can be optionally unsaturated and/or substituted with one or more "alkyl group substituents.” There can be optionally inserted along the alkylene group one or more oxygen, sulfur or substituted or unsubstituted nitrogen atoms (also referred to herein as "alkylaminoalkyl”), wherein the nitrogen substituent is alkyl as previously described.
  • An alkylene group can have about 2 to about 3 carbon atoms and can further have 6-20 carbons.
  • acyl refers to an organic carboxylic acid group wherein the -OH of the carboxyl group has been replaced with another substituent.
  • acyl specifically includes arylacyl groups, such as an acetylfuran and a phenacyl group. Specific examples of acyl groups include acetyl and benzoyl.
  • Cyclic and “cycloalkyl” refer to a non-aromatic mono- or multicyclic ring system of about 3 to about 10 carbon atoms, e.g., 3, 4, 5, 6, 7, 8, 9, or 10 carbon atoms.
  • the cycloalkyl group can be optionally partially unsaturated.
  • the cycloalkyl group also can be optionally substituted with an alkyl group substituent as defined herein, oxo, and/or alkylene.
  • cyclic alkyl chain There can be optionally inserted along the cyclic alkyl chain one or more oxygen, sulfur or substituted or unsubstituted nitrogen atoms, wherein the nitrogen substituent is hydrogen, alkyl, substituted alkyl, aryl, or substituted aryl, thus providing a heterocyclic group.
  • Representative monocyclic cycloalkyl rings include cyclopentyl, cyclohexyl, and cycloheptyl.
  • Multicyclic cycloalkyl rings include adamantyl, octahydronaphthyl, decalin, camphor, camphane, and noradamantyl.
  • Alkoxyl refers to an alkyl-O- group wherein alkyl is as previously described.
  • alkoxyl as used herein can refer to, for example, methoxyl, ethoxyl, propoxyl, isopropoxyl, butoxyl, f-butoxyl, and pentoxyl.
  • oxyalkyl can be used interchangably with “alkoxyl”.
  • Aryloxyl refers to an aryl-O- group wherein the aryl group is as previously described, including a substituted aryl.
  • aryloxyl as used herein can refer to phenyloxyl or hexyloxyl, and alkyl, substituted alky!, halo, or alkoxyl substituted phenyloxyl or hexyloxyl.
  • Alkyl refers to an aryl— alkyl— group wherein aryl and alkyl are as previously described, and included substituted aryl and substituted alkyl.
  • exemplary aralkyl groups include benzyl, phenylethyl, and naphthylmethyl.
  • Aralkyloxyl refers to an aralkyl-O- group wherein the aralkyl group is as previously described.
  • An exemplary aralkyloxyl group is benzyloxyl.
  • Ethers are groups of the structure -R-O-R, wherein each R is alkyl, substituted alkyl, aryl, aralkyl, or substituted aryl.
  • amine refers to NH 3 , primary amines (i.e., compounds of the structure H 2 NR, wherein R is alkyl, substituted alkyl, aryl, aralkyl, or substituted aryl), secondary amines (i.e., compounds of the structure HNR 2 , wherein each R is independently alkyl, substituted alkyl, aryl, aralkyl, or substituted aryl or wherein the two R groups together are an alkylene group), and tertiary amines (i.e., compounds of the structure NR 3 , wherein each R is independently alkyl, substituted alkyl, aryl, aralkyl, or substituted aryl or wherein two R groups together are an alkylene group).
  • primary amines i.e., compounds of the structure H 2 NR, wherein R is alkyl, substituted alkyl, aryl, aralkyl, or substituted aryl
  • secondary amines
  • amine also refers to aromatic amines, including aromatic compounds having a nitrogen atom in the aromatic ring, such as, but not limited to, pyridine, bipyridines, indole, pyrimidine, and the like, as well as amines with aromatic substituents (e.g., aniline).
  • hydroxyl and "hydroxyl” refer to the -OH group.
  • hydroxyalkyl or “hydroxy-substituted alkyl” refer to an alkyl group substituted with an -OH group.
  • mercapto or “thiol” refer to the -SH group.
  • a “thioether” refers to a -R-S-R group wherein each R is independently alkyl or aryl.
  • oxo refers to a compound described previously herein wherein a carbon atom is replaced by an oxygen atom.
  • alkene refers to a group having a carbon-carbon double bond.
  • alkyne refers to a group having a carbon-carbon triple bond.
  • sil refers to groups comprising silicon atoms (Si).
  • silicoxy and sil ether refer to groups or compounds including a silicon-oxygen (Si-OR) bond. In some embodiments, the terms refer to compounds comprising one, two, three, or four alkoxy, aralkoxy, or aryloxy groups bonded to a silicon atom. Each alkyloxy, aralkoxy, or aryloxy group can be the same or different.
  • sianol refers to the Si-OH group.
  • poly(siloxane) refers to a polymeric group of the formula R 2 SiO, wherein R is H, alkyl, aralkyl, or aryl.
  • poly(silsesquioxane) refers to a polymeric group of the formula RSiOi .5 , wherein R is H, alkyl, aralkyl, or aryl.
  • diaqua complex refers to a metal coordination complex comprising two water ligands.
  • nonplatinum an atom, chemical moiety, metal complex, or molecule that does not contain a platinum atom.
  • hydrophilic refers to the ability of a molecule or chemical species to interact with water. Thus, hydrophilic molecules are typically polar or have groups that can hydrogen bond to water.
  • hydrophobic refers to a molecule that interacts poorly with water (e.g., does not dissolve in water or does not dissolve in water to a large extent).
  • lipophilic refers to a molecule or chemical species that interacts (e.g., dissolves in) fat or lipids.
  • amphiphilic refers to a molecule or species that has both hydrophilic and hydrophobic (or lipophilic) attributes.
  • Nanoscale coordination polymers are a class of materials constructed from metal complexes having polydentate bridging ligands that can form coordination bonds to metals in more than one metal complex. Due to their inherent solubility in aqueous environments, NCPs can be designed to facilitate the controlled release of therapeutic groups, such as anticancer therapeutic agents. For example, anticancer therapeutic agents or their prodrugs can be bridging ligands in coordination polymers that make up the NCPs or can be the metal complexes, themselves.
  • platinum-based anticancer prodrugs i.e., cisplatin, carboplatin, and oxaliplatin
  • Figure 1 Some examples of platinum-based anticancer prodrugs, i.e., cisplatin, carboplatin, and oxaliplatin, are shown in Figure 1. They are used for treating testicular, ovarian, bladder, head and neck, esophageal, small and non-small cell lung, breast, cervical, stomach and prostate cancers, as well as Hodgkin's and non-Hodgkin's lymphomas, neuroblastoma, sarcomas, multiple myeloma, melanoma, and mesothelioma. These platinum-containing anticancer prodrugs convert to the highly potent platinum-aqua complexes inside cells.
  • the prodrugs currently used are not specific to cancer cells and can taken up by noncancerous cells, leading to many severe side effects including nausea and vomiting, kidney toxicity, blood test abnormalities, low white blood cell count, low red blood cell count, peripheral neuropathy, hearing loss, and hair loss. Often these side effects limit the dose and, thus, the therapeutic efficiency of platinum-based anticancer drugs.
  • stabilized NCPs can be prepared which are designed for sustained release of anticancer platinum- based prodrugs upon delivery to an intended tissue or tissues.
  • the presently disclosed subject matter provides novel coordination polymers (and nanomaterials thereof) comprising coordination polymers that comprise a plurality of platinum metal complexes.
  • L organic bridging ligands
  • the coordination polymer nanoparticles can be prepared by the addition of an initiating (i.e., "poor") solvent to a precursor solution containing a c/s-(NR3)2PtL-2 complex, which can be dicationic or neutral depending on the charge carried by the L ligands, and the organic bridging ligands.
  • an initiating solvent i.e., "poor”
  • the nanoparticles can be obtained under reverse microemulsion conditions.
  • the bridging ligands can be further linked or crosslinked by a second metal center (i.e., M 3+ or M 2+ ) such as lanthanide (Ln 3+ ) or a non-platinum transition metal (e.g., Zn 2+ ) to form a bimetallic coordination polymer.
  • the coordination polymers can comprise a plurality of Pt(II) metal complexes wherein the plurality of metal complexes comprise two or more different Pt(II) metal compexes (e.g., two or more different Pt(II) anticancer prodrugs).
  • Pt(IV) complexes can also be used as prodrugs for cancer therapy. These Pt(IV) complexes can be readily reduced to Pt(II) species such as cisplatin and oxaliplatin under physiological conditions, and they can be further transformed to their corresponding highly cytotoxic Pt(II) bis(aqua) complexes. Pt(IV) complexes, such as c,c,f-Pt(NH 3 ) 2 CI 2 (O 2 CCH 2 CH 2 CO 2 H) 2 (i.e., disuccinatocisplatin (DSCP)) can be incorporated into coordination polymers.
  • DSCP disuccinatocisplatin
  • Pt(IV) metal complex-based coordination polymers can also be further linked or crosslinked by a second metal center such as a lanthanide metal (Ln ) or a non-platinum transition metal (e.g., Zn 2+ ) to form bimetallic coordination polymer nanoparticles.
  • a second metal center such as a lanthanide metal (Ln ) or a non-platinum transition metal (e.g., Zn 2+ ) to form bimetallic coordination polymer nanoparticles.
  • a second metal center such as a lanthanide metal (Ln ) or a non-platinum transition metal (e.g., Zn 2+ ) to form bimetallic coordination polymer nanoparticles.
  • a second metal center such as a lanthanide metal (Ln ) or a non-platinum transition metal (e.g., Zn 2+ ) to form bimetallic coordination polymer nanoparticles.
  • the coordination polymers or their NCPs can comprise a plurality of Pt(II) metal complexes wherein the plurality of metal complexes comprise two of more different Pt(II) metal compexes Further, as shown in Figure 4, coordination polymers and NCPs of the presently disclosed subject matter can comprise a mixture of Pt(II) and Pt(IV) metal complexes.
  • the presently disclosed subject matter provides a nanoparticle comprising a coordination polymer comprising a plurality of platinum metal complexes wherein one or more of the platinum metal complexes each comprises: a platinum metal atom; at least one nonbridging ligand, wherein the at least one nonbridging ligand is bonded to the platinum metal atom through at least one coordination bond; and at least one bridging ligand, wherein the at least one bridging ligand is bonded to the platinum metal atom through at least one coordination bond and comprises at least one linking moiety, wherein each of the at least one linking moiety is bonded to an additional metal atom via a coordination bond.
  • the presently disclosed subject matter provides nanoparticles comprising coordination polymers that act as prodrugs of platinum-based anti-cancer therapeutics.
  • the coordination polymers "dissolve" (i.e., one or more ligands of individual platinum metal complexes can be replaced by wafer). If the ligands replaced by water include the bridging ligands, one or more of the individual platinum metal complexes are released from the polymer, either in a biologically active form, or in a form which can be further reacted with water to provide a biologically active (e.g., cytotoxic) form of the complex (e.g., a Pt(II) bis(aqua) species).
  • a biologically active e.g., cytotoxic
  • linking moieties of the bridging ligands can include any group that forms a coordination bond with a metal atom.
  • linking moieties can include, but are not limited to carboxylates, carboxylic acids, esters, amides, carbonates, carbamates, amines (e.g., primary amines, secondary amines or tertiary amines), hydroxyls (e.g., hydroxy-substituted alkyls or hydroxy-substituted aryls), thiols, thioethers, phosphates, phosphonates, and ethers.
  • each linking moiety present in a bridging ligand is independently selected from the group consisting of a carboxylate, a carboxylic acid, an amine, and an amide.
  • Each of the at least one bridging ligand is bonded via one or more coordination bond to the platinum metal atom of one of the plurality of platinum metal complexes in the coordination polymer and to one additional metal atom, which can be the platinum metal atom of a second platinum metal complex in the coordination polymer.
  • the same bridging ligand can be part of at least two of the plurality of platinum metal complexes.
  • the additional metal atom can be a nonplatinum metal atom (e.g., a lanthanide, actinide, or nonplatinum transition metal atom).
  • the bridging ligands serve to link and/or crosslink the monomer units (i.e., the individual platinum metal complexes) of the coordination polymer to one another, either directly or through a second metal atom.
  • Bridging ligands can be nonpolymeric or polymeric.
  • the coordination polymer of the presently disclosed subject matter comprises only nonpolymeric bridging ligands.
  • nonpolymeric bridging ligand it is meant a bridging ligand having a formula:
  • nonpolymeric bridging ligands can be based on organic small molecules (i.e., molecules having a molecular weight (MW) of less than about 1000, 900, 800, 700, 600, or 500 daltons).
  • nonpolymeric bridging ligands can also include synthetic organic or natural product-based anticancer drugs.
  • the at least one bridging ligand is monodentate with respect to one or more metal atoms to which it is complexed.
  • the bridging ligand can be bonded to the platinum metal atom of a particular platinum metal complex through one coordination bond and comprises at least one linking moiety for bonding to an additional metal atom.
  • Each of the at least one linking moieties can be bonded to a single additional metal atom (e.g., the platinum metal atom of a second platinum metal complex or a nonplatinum metal atom).
  • the bridging ligand comprises at least two carboxylate groups, e.g., wherein one carboxylate group is involved in a coordination bond with the platinum metal complex and the second carboxylate group is involved in a coordination bond with the additional metal atom.
  • the bridging ligand can further comprise one or more additional moieties that are capable of bonding to additional metal atoms via coordination bonds, but which do not necessarily need to be involved in such bonds in the coordination polymer as formed.
  • suitable bridging ligands include not only dicarboxylates, but also tri-, tetra-, and penta-carboxylates as well.
  • Such multi- carboxylate bridging ligands include, but are not limited to, 1 ,4-benzene dicarboxylate (i.e., terephthalate); 1 ,3,5-benzene tricarboxylate, succinate, oxalate, malonate, succinate, glutarate, phthalate, isophthalate, citrate, isocitrate, propane-1 ,2,3-tricarboxylate, ethylene diamine tetraacetate, and the like.
  • 1 ,4-benzene dicarboxylate i.e., terephthalate
  • 1 ,3,5-benzene tricarboxylate succinate, oxalate, malonate, succinate, glutarate, phthalate, isophthalate, citrate, isocitrate, propane-1 ,2,3-tricarboxylate, ethylene diamine tetraacetate, and the like.
  • a platinum metal complex can comprise two bridging ligands selected from the group consisting of 1 ,4-benzene dicarboxylate (i.e., terephthalate); 1 ,3,5-benzene tricarboxylate, succinate, oxalate, malonate, succinate, glutarate, phthalate, isophthalate, citrate, isocitrate, ethylene diamine tetraacetate, diethylene triamine pentaacetic acid, diethylenetriamine tetraacetic acid, and propane-1 ,2,3-tricarboxylate.
  • 1 ,4-benzene dicarboxylate i.e., terephthalate
  • 1 ,3,5-benzene tricarboxylate succinate, oxalate, malonate, succinate, glutarate, phthalate, isophthalate
  • citrate isocitrate
  • ethylene diamine tetraacetate diethylene triamine pentaacetic acid
  • the bridging ligand is a benzene dicarboxylate (BDC; e.g., terephthalate, phthalate, isophthalate) or a benzene tricarboxylate (BTC; e.g., 1 ,3,5-benzene tricarboxylate).
  • BDC benzene dicarboxylate
  • BTC benzene tricarboxylate
  • the at least one bridging ligand can have mixed functionality.
  • the bridging ligand can be of the formula:
  • Cm-X-Lm wherein Cm is a moiety involved in a coordination bond with the platinum metal atom, Lm is the linking moiety, and X is a bivalent group covalent bonded to both Cm and Lm, wherein Cm and Lm are different types of chemical groups.
  • one of Cm and Lm can be an amine group and the other of Cm and Lm can be carboxylate, carboxylic acid, amide, hydroxyl, or ester.
  • both Cm and Lm can be amine or both of Cm and Lm can be of the same functionality where the functionality is other than carboxylate or amine.
  • the bridging ligand can be multi-dentate (e.g., bidentate) with respect to one or more metal atoms to which it is coordinated.
  • a platinum metal complex can comprise a single bridging ligand, wherein the single bridging ligand is bonded to the platinum metal atom through two coordination bonds and comprises two linking moieties (e.g. each of which is bonded to a different additional metal atom).
  • the bridging ligand is a bipyridine dicarboxylate.
  • the bipyridine dicarboxylate is selected from the group consisting of 2,2'-bipyridine-5,5'-dicarboxylate and 2,2'-bipyridine-4,4'-dicarboxylate.
  • Nonbridging ligands are ligands that are bonded via coordination bonds to the platinum metal atom of a single platinum metal complex.
  • nonbridging ligands are ligands that are not bonded to any additional metal atoms.
  • Suitable nonbridging ligands can be independently selected from the group including, but not limited to, NH 3 , primary amines, secondary amines, tertiary amines, diamines, halides (i.e., iodide, chloride, bromide, and fluoride), hydroxide, hydroxy-substituted alkyl groups, hydroxy-substituted aryl groups, esters, carboxylates, carboxylic acids, carbamates, thiols, amides, and combinations thereof.
  • Nonbridging ligands can be selected based on the ligands of platinum-based anticancer therapeutics known in the art, such as, but not limited to carboplatin, cisplatin, nedaplatin, oxaliplatin, and satraplatin.
  • the nonbridging ligands are selected from the group consisting of Cl, NH 3 , cyclohexylamine, acetate, hydroxyacetate, cyclohexanediamine, bipyridine, oxylate, malonate, and cyclobutane-1 ,1- dicarboxylic acid.
  • Nonbridging Iigands can be monodentate or multidentate (e.g., bidentate).
  • each platinum metal complex can comprise between 1 and 4 nonbridging Iigands (i.e., 1 , 2, 3, or 4 nonbridging Iigands), such that the platinum metal atom is involved in a total of 4 or 6 coordination bonds with the bridging and nonbridging Iigands of the complex.
  • each of the plurality of platinum metal complexes is independently selected from the group consisting of: Pt[(NH 3 ) 2 (CI) 2 (O 2 CCH 2 CH 2 CO 2 ) 2 ] (i.e., DSCP); Pt[(NH 3 ) 2 (CI) 2 ⁇ O 2 CC 6 H 3 (CO 2 ) 2 ⁇ 2 ]; dichloro(2,2'-bipyridine-4,4'-dicarboxylato)platinum (II); dichloro(2,2'-bipyridine-5,5'-dicarboxylato)platinum (II); and Pt[(NH 3 ) 2 (CI) 2 (ethylene diamine tetraacetate) ⁇ .
  • Pt[(NH 3 ) 2 (CI) 2 (O 2 CCH 2 CH 2 CO 2 ) 2 ] i.e., DSCP
  • the nanoparticles of the presently disclosed subject matter generally have at least one dimension less than about 1000 nm.
  • the nanoparticles can have at least one dimension less than about 500 nm, less than about 400 nm, less than about 300 nm, or less than about 200 nm.
  • the nanoparticles are approximately spherical and each have a diameter of about 500 nm or less.
  • the nanoparticles can each have a diameter of between about 20 nm and about 250 nm.
  • each nanoparticle can have a diameter of about 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, or 250 nm. In some embodiments, each nanoparticle has a diameter of between about 40 nm and about 70 nm.
  • anticancer drugs do not contain a metal, or do not contain a platinum metal atom, but do bear functional groups that can coordinate to various metal centers.
  • organic (i.e., non- metallic) anticancer drugs are shown in Figure 5.
  • metal centers e.g., either a platinum metal atom or a nonplatinum metal atom
  • these nonplatinum anticancer drugs or their prodrugs can be linked or crosslinked by the metal atoms to form polymeric species based on metal- ligand coordination bonds.
  • the presently disclosed subject matter provides coordination polymers comprising nonplatinum (e.g., small molecule, organic) anticancer drugs or prodrugs that are linked or crosslinked via platinum or nonplatinum metal atoms.
  • the presently disclosed subject matter provides coordination polymers and nanoparticles comprising coordination polymers that comprise both platinum- based anticancer prodrugs and nonplatinum anticancer drugs or prodrugs.
  • the potential synergistic anticancer effects of the platinum-based anticancer drugs and the nonplatinum anticancer drugs can add to the potency of the chemotherapy and can potentially alleviated the acquired drug resistance problem often faced by conventional anticancer drugs.
  • a nonplatinum (e.g., small molecule, organic) anticancer drug or prodrug can be used as a bridging ligand in a NCP comprising one or more platinum metal complex. Any nonplatinum anticancer drug or prodrug known in the art and having suitable functionality for coordinating to metal atoms can be used.
  • Suitable nonplatinum anticancer drug bridging ligands include, but are not limited to, methotrexate, folic acid, leucovorin, vinblastine, vincristine, melphalan, imatinib, pemetrexed, vindesine, anastrozole, doxorubicin, cytarabine, azathioprine, letrozole and carboxylates thereof.
  • one or more nonplatinum anticancer drugs or prodrugs can be grafted onto the surface of a NCPs (e.g., a NCPs comprising a plurality of platinum metal complexes).
  • the nonplatinum anticancer drugs can be grafted onto the surface of the NCPs via covalent linkages that are cleavable under biological conditions (e.g., at a certain pH or in response to a reductant present in vivo and/or in vitro). Such linkages can be enzymatically cleavable.
  • the nonplatinum anticancer therapeutic can be derivatized (e.g., covalently bonded) with a polymeric or oligomeric linker group (e.g., a siloxane or poly(ethylene glycol)) that can be used as a chemical tether between the drug and the nanoparticle.
  • a polymeric or oligomeric linker group e.g., a siloxane or poly(ethylene glycol)
  • the presently disclosed subject matter provides a nanoscale coordination polymer that comprises anticancer drugs or prodrugs and is free of platinum.
  • the presently disclosed subject matter provides a coordination polymer comprising a plurality of nonplatinum metal complexes wherein the nonplatinum metal complexes are linked via bridging ligands that comprise nonplatinum anticancer drugs.
  • the nonplatinum anticancer bridging ligands can comprise known organic anticancer drugs, such as, but not limited to those, shown in Figure 5, or any other known anticancer drug that comprises chemical groups capable of forming coordination bonds with metal atoms.
  • the nonplatinum metal complexes can comprise any suitable metal atom, e.g., a nonplatinum transition metal atom, a lanthanide metal atom, or an actinide metal atom.
  • Pt(II) and/or Pt(IV) prodrugs can be polymerized with polymers or polymerizable monomers (i.e., represented by the oval shape in the uppermost reaction shown in Figure 6) to form copolymers of the coordination metal complexes and other polymers.
  • the Pt(II) or R(IV) prodrugs used in the preparation of NCPs having polymeric bridging ligands comprise a prelinking moiety that has a chemical functional group that can react with a polymerizable monomer. See Figure 6, middle reaction scheme.
  • Such prelinking moieties include siloxyl functional groups that can react with silyl ethers, poly(siloxanes) or poly(silsesquioxanes).
  • the Pt(II) or Pt(IV) prodrug contains a functional group (e.g., an alkene) that can react with acrylic acid or acrylamide to form poly(acrylate), poly(acrylamide) or other organic polymer- based linking moieties. See Figure 6, lower reaction scheme.
  • one or more bridging ligand in an NCP can be polymeric.
  • polymeric bridging ligand is meant a bridging ligand of the formula:
  • Cm-[X] n -Lm wherein Cm is a moiety involved in a coordination bond with the platinum metal atom, Lm is a linking moiety, n is an integer greater than 1 , and X is a nonmethylene bivalent moiety. In some embodiments, X is a non-metallic moiety other than methylene. In some embodiments, X comprises a non- platinum metal atom. In some embodiments, n is greater than 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100. Typically, polymeric bridging ligands can have MWs greater than about 1000. In some embodiments, the polymeric briding ligand comprises poly(siloxane), poly(silsesquioxane), poly(acrylate) or poly(acrylamide).
  • the presently disclosed NCPs can include one or more components in addition to the platinum metal complexes, the small molecule nonplatinum cancer drugs or prodrugs, or the combination of platinum metal complexes and small molecule nonplatinum anticancer drugs or prodrugs.
  • additional components include photosensitizers, radiosensitizers, radionuclides, imaging agents, passivating agents, stabilizing agents, and targeting agents.
  • additional components can be incorporated into the nanoparticles as ligands of metal complexes, incorporated into pores in the nanoparticles (e.g., physically trapped in pores via co- precipitation during formation of the nanoparticles, wherein the components are not covalently bound to any metal complex), covalently linked to ligands of metal complexes, or linked via coordination bonds to the platinum or other metals present in the NCPs.
  • the additional components are grafted (i.e., chemically linked via covalent or coordination bonds) to an outer surface of the NCPs.
  • the additional components can include small molecule-based or polymer-based linkers in order to facility grafting or other incorporation of the additional compentent into the NCPs.
  • metal-containing imaging agents or radiotherapeutic agents can be used to provide the metal atom of a nanoscale coordination polymer.
  • an imaging agent or radiotherapeutic agent can be copolymerized with platinum-based anticancer prodrugs and/or small molecule, nonplatinum anticancer drugs or prodrugs. JlL Nanoscale Coordination Polymers for Combination Chemotherapeutic and Photodynamic Therapy
  • Some anticancer drugs require external stimuli such as intense laser light or X-ray radiation to render them cytotoxic.
  • photosensitizers can be used in combination with light at specific wavelengths to transform triplet state oxygen present in tissues into singlet state oxygen, which can react with nearby biomolecules.
  • PHOTOFRIN® i.e., porfimer sodium; Axcan Pharma PDT, Inc., Birmingham, Alabama, United States of America
  • PDT photodynamic therapy
  • the chemical structure of PHOTOFRIN® is shown in Figure 7.
  • the efficacy of the PDT approach to cancer therapy is limited by the lack of efficient methods for accumulating adequate doses of the photosensitizer in the tumor.
  • Other photosensitizers include verteprofin, the chlorins, and 5-aminolevulinic acid.
  • the NCPs can be prepared to include a photosensitizer and be used in conjunction with PDT.
  • the NCP can be prepared to include the photosensitizer copolymerized with platinum metal complexes.
  • the photosensitizer can be grafted to the surface of an NCP.
  • Radiosensitizers are compounds that can make tumor cells more sensitive to radiation therapy.
  • Metoxafin gadolinium XCYTRIN®; Pharmacyclics, Sunnyvale, California, United States of America
  • the structure of metoxafin gadolinium is also shown in Figure 7.
  • Motexafin gadolinium belongs to the texafrin family of expanded porphyrins. Because of its strong resemblance to porphyrins and other naturally occurring tetrapyrrolic prosthetic groups, the texaphyrins exhibit useful characteristics, such as favorable biolocalization on cancer cells.
  • texaphyrin family Two distinct differences of the texaphyrin family from porphyrins make them potentially useful as radiosensitizers.
  • the expanded and trianionic nature of the texaphyrin endows a much tighter coordination with large and trivalent lanthanide metals.
  • Ianthanide complexes of texaphyrins exhibit unique redox characteristics and act as powerful oxidizing agents (with a reduction potential Ey 2 of -0.041 for motexafin gadolinium). This latter property can make motexafin gadolinium an efficient X-ray radiation (XRT) enhancing agent (i.e., a radiosensitizer).
  • XRT X-ray radiation
  • the NCPs can be prepared to include a radiosensitizer and be used in conjunction with radiation therapy.
  • the NCP can be prepared to include a radiation source itself.
  • the presently disclosed NCPs can be prepared to include radionuclides (i.e., radioisotopes) that can release radiation upon delivery of the NCP to a targeted cell, tissue or organ.
  • a beta-emitting metal center (e.g., 99 Y) can be incorporated as a metal in a NCP either containing Pt-metal complexes or organic anticancer drugs, or both for use in combination chemotherapy and radiotherapy.
  • Nanoscale coordination polymer architecture allows for a large variety of contrast agents for different imaging modalities, including, but not limited to magnetic resonance imaging (MRI), optical imaging, positron emission tomography (PET) and single photon emission computed tomography
  • MRI magnetic resonance imaging
  • PET positron emission tomography
  • SPECT positron emission computed tomography
  • a paramagnetic metal atom such as the Ianthanide atom Gd 3+ can be incorporated into the particles (e.g., as an additional metal atom bound via coordination bonds to bridging ligands in bimetallic NCPs) so that the NCPs can be used as dual chemotherapeutic/MRI imaging agents.
  • Gd 3+ is often chosen as a metal atom for MRI contrast agents be.cause it has a very high magnetic moment and a symmetric electronic ground state. Transition metals, including, but not limited to, high spin Mn(II) and Fe(III) can also be incorporated into the NCPs to provide MRI contrast enhancement.
  • NCPs to the tumor sites can allow for high-resolution delineation of the tumors.
  • MR images taken pre- and post-administration of the NCPs can enable assessment of the effectiveness of the anticancer drugs or prodrugs.
  • an organic fluorophore e.g., a near-infrared dye
  • luminophore that has a chemical functional group or groups that coordinate to metal atoms
  • the resulting NCPs can be used as effecient optical imaging contrast agents.
  • radioactive metal centers can be doped into the NCPs for application for PET (e.g., when 64 Cu is the radioactive metal) or SPECT (e.g., when 111 In or 99m Tc is the radioactive metal).
  • 64 Cu can be added to a precursor solution containing non-radioactive transition metals and either Pt-containing or organic anticancer drugs to form an NCP.
  • 111 In and 99m Tc are doped into the NCPs containing Pt- containing or organic anticancer drugs. SPECT can then be used for real-time monitoring of the biodistribution of the NCPs in vivo.
  • NCPs are soluble in aqueous environments (e.g., biological environments)
  • a stabilizing agent can be a chemical group, molecule, or delivery vehicle (e.g., liposome or microemulsion) that shields the NCP from the aqueous environment, for example, by reducing or eliminating access of water molecules to the NCP, for a period of time.
  • the stabilizing agent can be biodegradable, either over time or upon contact with a chemical or biochemical agent.
  • the presently disclosed subject matter relates to nanoparticles comprising a core and an outer layer.
  • the core can comprise a coordination polymer.
  • the outer layer which can surround the core, can comprise an inorganic or organic shell.
  • the NCP can comprise a coordination polymer core and an outer shell comprising a metal oxide, a silica- based polymer (e.g., silica (SiOa), a poly(siloxane), or a poly(silsesquioxane)), an organic polymer (e.g., polyvinylpyrrolidone (PVP), a polyamide or a polyester), a lipid bilayer (e.g., a liposome), or combinations thereof.
  • a silica- based polymer e.g., silica (SiOa), a poly(siloxane), or a poly(silsesquioxane)
  • an organic polymer e.g., polyvinylpyrroli
  • the stabilizing layer can be further functionalized to impart biological compatibility (e.g., reduced immunogenicity or reduced biological clearance), muitimodality (e.g., use as a dual chemotherapeutic/imaging agent, a chemotherapeutic/MRI imaging/optical imaging agent, a dual chemotherapeutic/radiotherapeutic agent, dual chemotherapeutic/PDT agent, dual chemotherapeutic/radiosensitizing agent) and/or specificity (e.g., targeting to a specific type of cell, organ, or tissue in vivo and/or in vitro).
  • biological compatibility e.g., reduced immunogenicity or reduced biological clearance
  • muitimodality e.g., use as a dual chemotherapeutic/imaging agent, a chemotherapeutic/MRI imaging/optical imaging agent, a dual chemotherapeutic/radiotherapeutic agent, dual chemotherapeutic/PDT agent, dual chemotherapeutic/radiosensitizing agent
  • the outer layer and the core can be chemically bonded to one another (e.g., via one or more coordination bond or one or more covalent bonds). However, in some embodiments, the outer layer and the core are not chemically bonded to one another, rather, the outer surface or surfaces of the core are merely surrounded or encapsulated by the outer layer material.
  • Figure 8 shows an example of the synthesis of a stabilitzed NCP.
  • an core structure i.e., NCP-1 comprising a bimetallic coordination polymer is prepared from Pt[(NH3)2(CI)2(O2CCH2CH2CO2)2 ⁇ and Tb 3+ .
  • the core is then coated with PVP and silica (i.e., from the polymerization of tetraethyl orthosilicate (TEOS)) to provide the silica-modified NCP (i.e., NCP- f).
  • TEOS tetraethyl orthosilicate
  • the presently disclosed NCPs can be functionalized with biocompatible passivating molecules to deter the adsorption of plasma proteins and/or recognition by biological defense systems, such as the reticuloendothelial system (RES).
  • the NCP comprises a passivating moiety comprising a polyethylene glycol (PEG)-based polymer.
  • PEG polymers are widely commercially available (e.g., from Aldrich Chemical Company, Milwaukee, Wisconsin, United States of America) in a variety of sizes and with a variety of terminal functionalities to aid in their covalent attachment to the presently disclosed NCPs.
  • PEG is generally hydrophilic, non-biodegradable, and non- immunogenic.
  • the PEG can be any size (e.g., MW of about 500, 1000, 5000, 10,000, 25,000, or more) or polydispersity.
  • the PEG- based polymer is polyethylene oxide (PEO)-500.
  • the PEG is grafted onto the outer shell of a stabilized NCP or onto an outer surface of a non-stabilized NCP.
  • Other polymers that can be used as passivating agents include, but are not limited to, lipid bilayers (such as liposomes).
  • the NCP can comprise a targeting agent to direct the NCP, once administered, to a target cell, organ, or tissue.
  • the target is a diseased cell (i.e., a cell associated with at particular disease state, such as a cancer cell).
  • Any targeting moiety known to be located on the surface of the target diseased cells, or expressed by the diseased cells finds use with the presently disclosed particles.
  • an antibody directed against a cell surface moiety can be used.
  • the targeting moiety can be a ligand directed to a receptor present on the cell surface or vice versa.
  • targeting moieties include small molecules, peptides, and proteins (including antibodies or antibody fragments (e.g., FABs)).
  • Targeting moieties for use in targeting cancer cells can be designed around tumor specific antigens including, but not limited to, carcinoembryonic antigen, prostate specific antigen, tyrosinase, ras, HER2, erb, MAGE-1 , MAGE- 3, BAGE, MN, gplOO, gp75, p97, proteinase 3, a mucin, CD81 , CID9, CD63; CD53, CD38, CO-029, CA125, GD2, GM2 and O-acetyl GD3, M-TAA, M-fetal or M-urinary find use with the presently disclosed subject matter.
  • tumor specific antigens including, but not limited to, carcinoembryonic antigen, prostate specific antigen, tyrosinase, ras, HER2, erb, MAGE-1 , MAGE- 3, BAGE, MN, gplOO, gp75, p97, proteinase 3,
  • the targeting moiety can be designed around a tumor suppressor, a cytokine, a chemokine, a tumor specific receptor ligand, a receptor, an inducer of apoptosis, or a differentiating agent.
  • the targeting moiety can be developed to target a factor associated with angiogenesis.
  • the targeting moiety can be designed to interact with known angiogenisis factors such as vascular endothelial growth factor (VEGF). See Brannon-Peppas, L. and Blanchette, J. Q., Advanced Drug Delivery Reviews, 56, 1649-1659 (2004).
  • VEGF vascular endothelial growth factor
  • Tumor suppressor proteins provided for targeting include, but are not limited to, p16, p21 , p27, p53, p73, Rb, Wilms tumor (WT-1), DCC, neurofibromatosis type 1 (NF-1), von Hippel-Lindau (VHL) disease tumor suppressor, Maspin, Brush-1, BRCA-1 , BRCA-2, the multiple tumor suppressor (MTS), gp95/p97 antigen of human melanoma, renal cell carcinoma-associated G250 antigen, KS 1/4 pan-carcinoma antigen, ovarian carcinoma antigen (CA125), prostate specific antigen, melanoma antigen gp75, CD9, CD63, CD53, CD37, R2, CD81 , CO029, TM 1 L6 and SAS.
  • WT-1 Wilms tumor
  • DCC neurofibromatosis type 1
  • VHL von Hippel-Lindau
  • MTS multiple tumor suppressor
  • targeting is directed to factors expressed by an oncogene.
  • oncogene include, but are not limited to, tyrosine kinases, both membrane-associated and cytoplasmic forms, such as members of the Src family, serine/threonine kinases, such as Mos, growth factor and receptors, such as platelet derived growth factor (PDDG), SMALL GTPases (G proteins) including the ras family, cyclin-dependent protein kinases (cdk), members of the myc family members including c-myc, N-myc, and L-myc and bcl-2 and family members.
  • PDDG platelet derived growth factor
  • SMALL GTPases G proteins
  • cdk cyclin-dependent protein kinases
  • members of the myc family members including c-myc, N-myc, and L-myc and bcl-2 and family members.
  • Cytokines that can be targeted by the presently disclosed particles include, but are not limited to, IL-1 , IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL- 10, ILA 1 , IL-12, IL-13, IL-14, IL-15, TNF, GM-CSF, ⁇ -interferon, and ⁇ - interferon.
  • Chemokines that can be used include, but are not limited to, M1 P1 ⁇ , M1 P1 ⁇ , and RANTES.
  • Enzymes that can be targeted include, but are not limited to, cytosine deaminase, hypoxanthine-guanine phosphoribosyltransferase, galactose-1- phosphate uridyltransferase, phenylalanine hydroxylase, glucocerbrosidase, sphingomyelinase, ⁇ -L-iduronidase, glucose-6-phosphate dehydrogenase, HSV thymidine kinase, and human thymidine kinase.
  • Receptors and their related ligands that find use in the context of the presently disclosed subject matter include, but are not limited to, the folate receptor, adrenergic receptor, growth hormone receptor, luteinizing hormone receptor, estrogen receptor, epidermal growth factor(EGF) receptor, fibroblast growth factor receptor (FGFR), and the like.
  • EGF is overexpressed in brain tumor cells and in breast and colon cancer cells.
  • the targeting moiety is selected from the group consisting of folic acid, guanidine, transferrin, carbohydrates and sugars.
  • the targeting mofety is a peptide selected from the group consisting of the amino acid sequence RGD and TAT peptides.
  • the targeting moiety can comprise cyclic(RGDfK) (i.e., c(RGDfK)).
  • c(RGDfK) cyclic(RGDfK)
  • a suitably dervitized c(RGDfK) e.g., a siloxy-functionalized c(RGDfK)
  • the targeted NCP can slowly leach out the platinum metal complex after being taken up by a tumor that recognizes the RGD motif. See Scheme 8, final step.
  • Hormones and their receptors include, but are not limited to, growth hormone, prolactin, placental lactogen, luteinizing hormone, folicle-stimulating hormone, chorionic gonadotropin, thyroid-stimulating hormone, leptin, adrenocorticotropin (ACTH), angiotensin I, angiotensin IJ, ⁇ -endorphin, ⁇ - melanocyte stimulating hormone ( ⁇ -MSH), cholecystokinin, endothelin I 1 galanin, gastric inhibitory peptide (GIP), glucagon, insulin, amylin, lipotropins, GLP-1 (7-37) neurophysins, and somatostatin.
  • growth hormone prolactin, placental lactogen, luteinizing hormone, folicle-stimulating hormone, chorionic gonadotropin, thyroid-stimulating hormone, leptin, adrenocorticotropin (ACTH), angiotensin
  • vitamins both fat soluble and non-fat soluble vitamins placed in the targeting component of the nanomaterials can be used to target cells that have receptors for, or otherwise take up these vitamins.
  • Particularly preferred for this aspect are the fat soluble vitamins, such as vitamin D and its analogues, Vitamin E, Vitamin A, and the like or water soluble vitamins such as Vitamin C, and the like.
  • Antibodies can be generated to allow for the targeting of antigens or immunogens (e.g., tumor, tissue or pathogen specific antigens) on various biological targets (e.g., pathogens, tumor cells, and normal tissue).
  • the targeting moiety is an antibody or an antigen binding fragment of an antibody (e.g., Fab, F(ab')2, or scFV units).
  • antibodies include, but are not limited to polyclonal antibodies, monoclonal antibodies, chimeric antibodies, single chain antibodies, Fab fragments, and a Fab expression library. Other characteristics of the nanoparticle also can be used for targeting.
  • the enhanced permeability and retention (EPR) effect is used in targeting.
  • the EPR effect is the selective concentration of macromolecules and small particles in the tumor microenvironment, caused by the hyperpermeable vasculature and poor lymphatic drainage of tumors.
  • the exterior of the particle can be coated with or conjugated to a hydrophilic polymer to enhance the circulation half-life of the particle and to discourage the attachment of plasma proteins to the particle.
  • targeting agents can be chemically derivatized to incorporate chemical groups that can be used to graft or otherwise incorporate the targeting agents into NCPs.
  • the targeting agents can be covalently bound to coordination complexes that can be copolymerized into the NCPs.
  • the targeting agents can be derivatized with chemical groups that include functionalities that can react with stabilizing agents or layers surrounding NCPs.
  • compositions of the presently disclosed subject matter comprise in some embodiments a composition that includes a NCP and a pharmaceutically acceptable carrier.
  • Any suitable pharmaceutical formulation can be used to prepare the compositions for administration to a subject.
  • the composition and/or carriers can be pharmaceutically acceptable in humans.
  • suitable formulations can include aqueous and nonaqueous sterile injection solutions that can contain anti-oxidants, buffers, bacteriostatics, bactericidal antibiotics, and solutes that render the formulation isotonic with the bodily fluids of the subject; and aqueous and non-aqueous sterile suspensions that can include suspending agents and thickening agents.
  • the formulations can be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and can be stored in a frozen or freeze- dried (lyophilized) condition requiring only the addition of sterile liquid carrier, for example water for injections, immediately prior to use.
  • sterile liquid carrier for example water for injections, immediately prior to use.
  • Some exemplary ingredients are sodium dodecyl sulfate (SDS), in one example in the range of 0.1 to 10 mg/ml, in another example about 2.0 mg/ml; and/or mannitol or another sugar, for example in the range of 10 to 100 mg/ml, in another example about 30 mg/ml; and/or phosphate-buffered saline (PBS).
  • SDS sodium dodecyl sulfate
  • PBS phosphate-buffered saline
  • the presently disclosed NCPs can be formulated in liposomes or microemulsions by methods known in the art.
  • formulations of this presently disclosed subject matter can include other agents conventional in the art having regard to the type of formulation in question.
  • sterile pyrogen-free aqueous and non- aqueous solutions can be used.
  • compositions disclosed herein can be used on a sample either in vitro (for example, on isolated cells or tissues) or in vivo in a subject (i.e. living organism, such as a patient).
  • a subject i.e. living organism, such as a patient.
  • the subject is a human subject, although it is to be understood that the principles of the presently disclosed subject matter indicate that the presently disclosed subject matter is effective with respect to all vertebrate species, including warm-blooded vertebrates, such as mammals and birds, which are intended to be included in the terms "subject” and "patient".
  • a mammal is understood to include any mammalian species for which employing the compositions and methods disclosed herein is desirable, particularly agricultural and domestic mammalian species.
  • endangered such as Siberian tigers
  • economic importance animals raised on farms for consumption by humans
  • social importance animals kept as pets or in zoos
  • ruminants such as cattle, oxen, sheep, giraffes, deer, goats, bison, and camels
  • Bird subjects include those kinds of birds that are endangered, kept in zoos or as pets (e.g., parrots), as well as fowl, and more particularly domesticated fowl, for example, poultry, such as turkeys, chickens, ducks, geese, guinea fowl, and the like, as they are also of economic importance to humans.
  • domesticated swine pigs and hogs
  • ruminants horses, poultry, and the like.
  • Suitable methods for administering to a subject a composition of the presently disclosed subject matter include, but are not limited to, systemic administration, parenteral administration (including intravascular, intramuscular, intraarterial administration), oral delivery, buccal delivery, subcutaneous administration, inhalation, intratracheal installation, surgical implantation, transdermal delivery, local injection, and hyper-velocity injection/bombardment.
  • continuous infusion can enhance drug accumulation at a target site (see, for example, U.S. Patent No. 6,180,082).
  • the particular mode of drug administration used in accordance with the methods of the presently disclosed subject matter depends on various factors, including but not limited to the agent and/or carrier employed, the severity of the condition to be treated or treated and imaged, and mechanisms for metabolism or removal of the active agent following administration.
  • relatively superficial tumors can be injected intratumorally.
  • Internal tumors can be treated or treated and imaged following intravenous injection.
  • selective delivery of a composition to a target is accomplished by intravenous injection of the composition followed by hyperthermia treatment of the target.
  • compositions of the presently disclosed subject matter can be formulated as an aerosol or coarse spray.
  • Methods for preparation and administration of aerosol or spray formulations can be found, for example, in U.S. Patent Nos. 5,858,784;
  • an effective dose of a composition of the presently disclosed subject matter is administered to a subject or to a biological sample (e.g., a sample containing cancer cells).
  • An "effective amount” is an amount of the composition sufficient to produce an anticancer therapeutic effect (e.g., reduction of tumor size, reduction in cancer cell proliferation, cancer cell death, etc.).
  • Actual dosage levels of constituents of the compositions of the presently disclosed subject matter can be varied so as to administer an amount of the composition that is effective to achieve the desired effect for a particular subject and/or target. The selected dosage level can depend upon the activity of the composition and the route of administration.
  • the presently disclosed subject matter provides methods of treating cancer comprising the administration of the presently disclosed NCPs (e.g., NCPs comprising coordination polymers comprising a plurality of platinum metal complexes; NCPs comprising coordination polymers comprising anticancer therapeutic bridging ligands, or NCPs comprising coordination polymers comprising platinum metal complexes and anticancer therapeutic bridging ligands).
  • the NCP can be used in methods involving the treatment of cancer and the imaging of a cell, tissue, or organ.
  • the NCP can be used in a method of treating cancer that further comprises PDT or radiotherapy.
  • the NCP comprises a targeting agent and the method comprises delivery of the NCP to a specific cell, tissue or organ (e.g., a cell, tissue or organ associated with a particular disease, such as cancer).
  • the presently disclosed subject matter provides a method of inhibiting proliferation of a cancer cell, the method comprising contacting the cancer cell with a nanoparticle, wherein the nanoparticle comprises a coordination polymer comprising a plurality of platinum metal complexes.
  • inhibiting proliferation of a cancer cell is meant that contacting the cancer cell with the nanoparticle inhibits cell division of the cancer cell.
  • inhibiting proliferation can further comprise triggering apoptosis in the cancer cell or otherwise causing the death of the cancer cell.
  • the cancer cell can be any cancer cell.
  • Cancer cells treatable by the presently disclosed methods include, but are not limited to, skin cancer cells, connective tissue cancer cells, breast cancer cells, lung cancer cells, esophogeal cancer cells, stomach cancer cells, a head and neck cancer cell, pancreatic cancer cells, ovarian cancer cells, cervical cancer cells, uterine cancer cells, anogenital cancer cells, kidney cancer cells, bladder cancer cells, colon cancer cells, prostate cancer cells, retinal cancer cells, central nervous system cancer cells, and lymphoid cancer cells.
  • the cancer cell is selected from the group consisting of a breast cancer cell and a colon cancer cell.
  • the NCP can comprise a plurality of platinum (II) metal complexes, a plurality of platinum (IV) metal complexes, or a combination thereof.
  • One or more of the platinum metal complexes can comprise: a platinum metal atom; at least one nonbridging ligand, wherein the at least one nonbridging ligand is bonded to the platinum metal atom through at least one coordination bond; and at least one bridging ligand, wherein the at least one bridging ligand is bonded to the platinum metal atom through at least one coordination bond and comprises at least one linking moiety, wherein each of the at least one linking moiety is bonded to an additional metal atom via a coordination bond.
  • Suitable linking moieties include, but are not limited to, carboxylate, carboxylic acid, ester, carbamate, carbonate, amine (e.g., primary, secondary, tertiary, or aromatic amines), amide, hydroxyl, thiol, ether, thioether, phosphonate, and phosphate.
  • Nonbridging ligands can be independently selected from the group including, but not limited to, NH 3 , primary amines, secondary amines, tertiary amines, diamines, aromatic amines, halides, hydroxy-substituted alkyl, hydroxy-substituted aryl, esters, carboxylates, carboxylic acids, carbamates, amides, thiols, hydroxide, and combinations thereof.
  • the bridging ligand can comprise at least two carboxylate groups.
  • Bridging ligands can be either polymeric or nonpolymeric.
  • the NCP comprises only nonpolymeric bridging ligands.
  • the NCP can comprise one or more platinum metal complex(es) that comprise(s) two bridging ligands, where each of the two bridging ligands is bonded to the platinum metal atom through one coordination bond and comprises at least one linking moiety.
  • Each of the two bridging ligands can optionally be selected from a BDC or BTC.
  • each of the two bridging ligands can be independently selected from the group including but not limited to 1 ,4-benzene dicarboxylate; 1 ,3,5-benzene tricarboxylate; succinate; and ethylene diamine tetraacetate.
  • the NCP can comprise one or more platinum metal complex(es) that comprise(s) one bridging ligand, where the one bridging ligand is bonded to the platinum metal atom through two coordination bonds and comprises at least two linking moieties.
  • the bridging ligand is a bipyridine dicarboxylate, such as, but not limited to, 2,2'-bipy ⁇ idine-5,5'-dicarboxylate and 2,2'-bipyridine-4,4'-dicarboxylate.
  • the NCP comprises one or more platinum metal complex(es) that comprise(s) a bridging ligand where the bridging ligand is a nonplatinum anticancer drug or anticancer prodrug.
  • Suitable nonplatinum anticancer drug bridging ligands include, but are not limited to, methotrexate, folic acid, leucovorin, vinblastine, vincristine, melphalan, imatinib, pemetrexed, vindesine, anastrozole, doxorubicin, cytarabine, azathioprine, letrozole and carboxylates thereof.
  • the NCP can comprise at least one polymeric bridging ligand.
  • Polymeric bridging ligands can comprise polymers such as, but not limited to, poly(silsesquioxane), poly(siloxane), poly(acrylate) and poly(acrylamide).
  • the NCP is bimetallic and includes one or more additional nonplatinum metal atom in addition to the platinum metal atoms of the platinum metal complexes.
  • the nonplatinum metal atoms can include nonplatinum transition metal atoms, lanthanide metal atoms, and actinide metal atoms.
  • the NCP can be a bimetallic coordination polymer comprising formulas such as Tb x [Pt(L) n (BL) m ] y or Zn x [Pt(L) n (BL) m ]y, wherein each L is a nonbridging ligand and each BL is a bridging ligand.
  • each of the plurality of platinum metal complexes in the NCP can be independently selected from the group including but not limited to:
  • a stabilized NCP can be contacted with the cell.
  • the NCP can comprise a core and an outer layer, wherein the core comprises a coordination polymer and the outer layer surrounds the core and comprises a group that shields the NCP from the aqueous environment, either partially or totally, for a period of time.
  • the NCP can be used in methods where controlled release of platinum metal complexes and/or other anticancer therapeutics is desired.
  • the controlled release can relate to sustained release over a given period of time or to targeted release in the presence of a particular cell type or in the presence of particular conditions (e.g., pH, presence of a particular enzyme or biological reductant, etc).
  • the outer layer can comprise a metal oxide, an organic polymer (e.g., PVP), a silica-based polymer (e.g., SiO 2 ), a lipid bilayer, or a combination thereof.
  • the NCP can comprise one or more of a photosensitizer, a radiosensitizer, a radionuclide, an imaging agent, a passivating agent, and a targeting agent.
  • the NCP can comprise a photosensitizer and the method can further comprise administering light to the cancer cell (e.g., laser light of a particular wavelength or wavelength range) during or after contacting the cell with the NCP.
  • the NCP can comprise a radiosensitizer, and the method can further comprise administering radiation or a source of radiation to the cancer cell during or following the contacting of the cell with the NCP.
  • the NCP can comprise both a radiosensitizer and a radionuclide in addition to the platinum metal complexes and/or small molecule anticancer drugs and administering the NCP to the cell provides both chemotherapeutic treatment and radiation therapy.
  • the NCP can comprise a targeting agent that directs entry of the NCP or its components into the cell via receptor-mediated endocytosis.
  • the cell can comprise an angiongenic cancer cell and/or a cancer cell known to express an integrin receptor on its surface, and the NCP can comprise a targeting agent comprising the RGD tripeptide motif, such as a targeting agent comprising C(RGDfK).
  • the NCP can comprise one or more imaging agents.
  • the imaging agents can include, but are not limited to, optical imaging agents, MRI agents, PET agents, SPECT agents and combinations thereof.
  • the method further comprises imaging the cancer cell or the sample comprising the cancer cell via one or more suitable imaging technique.
  • the imaging can be performed prior to contacting the cell with the NCP, while contacting the cell with the NCP, after contacting the cell with the NCP, or any combination thereof.
  • the imaging can be performed, for example, to show entry of the NCP into one or more cancer cells, to show the location of the cancer cells in a larger sample (e.g., a tissue, organ, or subject), or to provide evidence of reduction of proliferation of the cancer cells via administration with the NCP.
  • the cancer cell is present in an in vitro sample (e.g., a cell culture, a cell culture extract, or an excised organ or tissue). In some embodiments, the cell is present in a subject.
  • an in vitro sample e.g., a cell culture, a cell culture extract, or an excised organ or tissue.
  • the cell is present in a subject.
  • the presently disclosed subject matter provides a method of treating cancer in a subject in need of treatment thereof, the method comprising administering to the subject a nanoparticle comprising a coordination polymer, wherein the coordination polymer comprises a plurality of platinum metal complexes.
  • treating cancer is meant providing the nanoparticle in an effective amount to prevent proliferation of the cancer (e.g., to prevent an increase in the size of a tumor or the number of cancer cells present in a subject or to prevent metastasis of the cancer to additional locales (e.g., organs or tissues) in the body of the subject), to reduce the size of a tumor related to the cancer or to otherwise reduce the number of cancer cells present in the subject, or to relieve one or more symptoms related to the presence of the cancer in the subject.
  • proliferation of the cancer e.g., to prevent an increase in the size of a tumor or the number of cancer cells present in a subject or to prevent metastasis of the cancer to additional locales (e.g., organs or tissues) in the body of the subject
  • additional locales e.g., organs or tissues
  • the cancer to be treated according to the presently disclosed methods can include any cancer, such as, but not limited to a skin cancer, a connective tissue cancer, an esophogeal cancer, a breast cancer, a lung cancer, a stomach cancer, a pancreatic cancer, an ovarian cancer, a cervical cancer, a uterine cancer, an anogenital cancer, a kidney cancer, a bladder cancer, a colon cancer, a prostate cancer, a retinal cancer, a central nervous system cancer, or a lymphoid cancer.
  • the cancer is breast cancer or colon cancer.
  • the subject is a mammal. In some embodiments, the subject is a human.
  • the nanoparticle is administered in a liposomal or microemulsion formulation. The nanoparticle can be administered via any suitable route.
  • the NCP is administered via an intravenous route or intratumoral route of administration.
  • the NCP can comprise a plurality of platinum (II) metal complexes, a plurality of platinum (IV) metal complexes, or a combination thereof.
  • One or more of the platinum metal complexes can comprise: a platinum metal atom; at least one nonbridging ligand, wherein the at least one nonbridging ligand is bonded to the platinum metal atom through at least one coordination bond; and at least one bridging ligand, wherein the at least one bridging ligand is bonded to the platinum metal atom through at least one coordination bond and comprises at least one linking moiety, wherein each of the at least one linking moiety is bonded to an additional metal atom via a coordination bond.
  • Suitable linking moieties include, but are not limited to, carboxylate, carboxylic acid, ester, carbamate, carbonate, amine (e.g., primary, secondary, tertiary, or aromatic amine), amide, hydroxyl, thiol, ether, thioether, phosphonate, and phosphate.
  • Nonbridging ligands can be independently selected from the group including, but not limited to, NH 3 , primary amines, secondary amines, tertiary amines, diamines, aromatic amines, halides, hydroxy-substituted alky!, hydroxy-substituted aryl, esters, carboxylates, carboxylic acids, carbamates, amides, thiols, hydroxide, and combinations thereof.
  • the bridging ligand can comprise at least two carboxylate groups.
  • Bridging ligands can be either polymeric or nonpolymeric.
  • the NCP comprises only nonpolymeric bridging ligands.
  • the NCP can comprise one or more platinum metal complex(es) that comprise(s) two bridging ligands, where each of the two bridging ligands is bonded to the platinum metal atom through one coordination bond and comprises at least one linking moiety.
  • Each of the two bridging ligands can be selected from a BDC or BTC.
  • each of the two bridging ligands can be independently selected from the group including but not limited to 1 ,4-benzene dicarboxylate; 1 ,3,5-benzene tricarboxylate; succinate; and ethylene diamine tetraacetate.
  • the NCP can comprise one or more platinum metal complex(es) that comprise(s) one bridging ligand, where the one bridging ligand is bonded to the platinum metal atom through two coordination bonds and comprises at least two linking moieties.
  • the bridging ligand is a bipyridine dicarboxylate, such as, but not limited to, 2,2'-bipyridine-5,5'-dicarboxylate and 2,2'-bipyridine-4,4'-dicarboxyiate.
  • the NCP comprises one or more platinum metal complex(es) that comprise(s) a bridging ligand where the bridging ligand is an nonplatinum anticancer drug or anticancer prodrug.
  • Suitable nonplatinum anticancer drug bridging ligands include, but are not limited to, methotrexate, folic acid, leucovorin, vinblastine, vincristine, melphalan, imatinib, pemetrexed, vindesine, anastrozole, doxorubicin, cytarabine, azathioprine, letrozole and carboxylates thereof.
  • the NCP can comprise at least one polymeric bridging ligand.
  • Polymeric bridging ligands can comprise polymers such as, but not limited to, poly(silsesquioxane), poly(siloxane), poly(acrylate) and poly(acrylamide).
  • the NCP is bimetallic and includes one or more additional nonplatinum metal atom in addition to the platinum metal atoms of the platinum metal complexes.
  • the nonplatinum metal atoms can include nonplatinum transition metal atoms, lanthanide metal atoms, and actinide metal atoms.
  • the NCP can be a bimetallic coordination polymer comprising formulas such as Tb x [Pt(L) n (BL) m ]y or Zn x [Pt(L)n(BL) m ] yi wherein each L is a nonbridging ligand and each BL is a bridging ligand.
  • each of the plurality of platinum metal complexes in the NCP can be independently selected from the group including but not limited to: Pt[(NH 3 ) 2 (CI) 2 (O 2 CCH 2 CH 2 COZ) 2 ];
  • a stabilized NCP can be administered to the subject.
  • the NCP can comprise a core and an outer layer, wherein the core comprises a coordination polymer and the outer layer surrounds the core and comprises a group that shields the NCP from the aqueous environment in vivo following administration, either partially or totally, for a period of time.
  • the NCP can be used in methods where controlled release of platinum metal complexes and/or other anticancer therapeutics is desired. The controlled release can relate to sustained release over a given period of time or to targeted release in the presence of a particular cell type or in the presence of particular conditions
  • the outer layer can comprise a metal oxide, an organic polymer (e.g., PVP), a silica-based polymer (e.g., SiO 2 ), a lipid bilayer, or a combination thereof.
  • PVP organic polymer
  • silica-based polymer e.g., SiO 2
  • lipid bilayer e.g., lipid bilayer, or a combination thereof.
  • the NCP further comprises one or more of a photosensitizer, a radiosensitizer, a radionuclide, a passivating agent, an imaging agent, and a targeting agent.
  • the NCP can comprise a photosensitizer and the method can further comprise administering an external stimulus (e.g., laser light of a particular wavelength or wavelength range) to the subject (e.g., to a tumor site within the subject) during or after administering the nanoparticle.
  • an external stimulus e.g., laser light of a particular wavelength or wavelength range
  • the nanoparticle can comprise a radiosensitizer, and the method can further comprise administering radiation or a source of radiation (e.g., a radionuclide) to the subject during or following administering the nanoparticle.
  • a source of radiation e.g., a radionuclide
  • the nanoparticle can comprise both a radiosensitizer and a radionuclide in addition to the platinum metal complexes and/or small molecule anticancer drugs and administering the nanoparticle to the cells provides both chemotherapeutic treatment and radiation therapy.
  • the radiation or radiation source comprises an external stimulus (e.g., X-ray radiation).
  • the NCP can comprise a targeting agent that directs delivery of the nanoparticle to a particular type of cell, organ or tissue within the subject.
  • the NCP can comprise a targeting agent specific for cancer cells or for a particular type of cancer cells.
  • the NCP can comprise a targeting agent comprising the RGD tripeptide motif, such as a targeting agent comprising c(RGDfK).
  • the NCP can comprise one or more imaging agents.
  • the imaging agents can include, but are not limited to, optical imaging agents, MRI agents, PET agents, SPECT agents and combinations thereof.
  • the method can further comprise imaging the comprising imaging delivery of the nanoparticle in one or more cells, tissues or organs in the subject following administration of the nanoparticle via one or more suitable imaging technique.
  • the imaging can be performed prior to, during, or after administration of the nanoparticle, or in any combination thereof.
  • the imaging can be performed, for example, to show entry of the nanoparticles into one or more cancer cells, to show the location and/or size of the cancer, or to provide evidence that administration of the nanoparticle provides effective treatment of the cancer.
  • the NCPs can be prepared via any suitable method.
  • the NCPs can be synthesized either via precipitation methods involving initiator solvents (also referred to as "poor" solvents) or via microemulsion techniques.
  • the precipitation methods comprise providing solutions of the components of the coordination polymers (e.g., the platinum metal complexes, bridging ligands, and/or additional metal atoms or components) in solvents in which the components are soluble (i.e., readily dissolve to provide a clear solution). Addition of a solvent or solvent mixture (i.e., the initiator or "poor" solvent) in which the components are not as soluble leads to polymerization of the components and precipitation of the resultant nanoparticles.
  • the components of the NCPs are provided in one or more microemulsion solutions. Contacting and/or stirring the microemulsion solutions leads to polymerization of the components and formation of the NCPs.
  • the presently disclosed subject matter provides a method of synthesizing a nanoparticle comprising a coordination polymer comprising a plurality of platinum metal complexes, wherein the method comprises: providing a solution comprising a first solvent, at least one bridging ligand precursor, and a plurality of platinum diaqua complexes selected from the group consisting of platinum (II) diaqua complexes, platinum (IV) diaqua complexes, and mixtures thereof; and adding a second solvent to the solution to precipitate the nanoparticle.
  • the method further comprises adjusting the pH of the solution prior to adding the second solvent.
  • a base e.g., NaOH, KOH, LiOH, or sodium carbonate
  • an aqueous solution of an acid e.g., HCI or sulfuric acid
  • Adjusting the pH can be performed, for instance, to increase the solubility of a bridging ligand precursor or of diaqua complexes in the solution.
  • the first solvent can be any solvent or solvent mixture which, when provided in a suitable volume, will dissolve an amount of bridging ligand precursor and diaqua complex sufficient for preparing a desired amount of NCP.
  • the first solvent can comprises water, dimethyl sulfoxide (DMSO), or a combination thereof.
  • the at least one bridging ligand precursor comprises a nonpolymeric molecule.
  • the at least one bridiging moiety can be a molecule such as, but not limited to, a benzene dicarboxylic acid, a benzene dicarboxylate, a carboxylate-substituted styrene, a carboxylate-substituted silyl ether, a bipyridine dicarboxylic acid, a bipyridine dicarboxylate, a dicarboxylic anhydride, a diacyl dichloride, and a nonplatinum anticancer drug.
  • Suitable nonplatinum anticancer drugs include, but are not limited to, methotrexate, folic acid, leucovorin, vinblastine, vincristine, melphalan, imatinib, pemetrexed, vindesine, anastrozole, doxorubicin, cytarabine, azathioprine, letrozole, and carboxylates thereof.
  • the second solvent should be a solvent that when mixed into the solution, decreases the solubility of one or more of the bridging ligand precursor, the platinum diaqua complexes, or the NCPs prepared therefrom.
  • the second solvent is selected from the group including but not limited to acetone, an alcohol, ether, acetonitrile, and mixtures thereof.
  • the solution can comprise one or more additional components.
  • the solution can further comprise metal compounds that contain nonplatinum metal atoms.
  • the nonplatinum metal atoms can be nonplatinum transition metal atoms, lanthanide metal atoms, actinide metal atoms, or combinations thereof.
  • the nonplatinum metal atom is Tb 3+ or Zn 2+ .
  • the solution can comprise a Tb-containing compound, such as, but not limited to, TbCI 3 .
  • the bridging ligand precursor is polymeric or comprises a polymerizable monomer.
  • Suitable polymerizable monomers can include, but are not limited to acrylic acid, acrylamide, and silyl ethers.
  • the solution can further comprise an additional component such as a radionuclide, an imaging agent, a photosensitizer, and a radiosensitizer, and adding the second solvent co-precipitates the additional component, thereby incorporating the additional component into the nanoparticle (e.g., as an additional metal atom in the coordination polymer if the additional component comprises a radionuclide, covalently bound to components of the coordination polymer, or physically sequestered in pores within the coordination polymer).
  • an additional component such as a radionuclide, an imaging agent, a photosensitizer, and a radiosensitizer
  • the NCPs can be prepared from microemulsions.
  • Microemulsions particularly, water-in-oil, or reverse, microemulsions have been used to synthesize a variety of nanophase materials such as organic polymers, semiconductor nanoparticles (see Xu and Akins, Material. Letters, 58, 2623 (2004)), metal oxides, and nanocrystals consisting of cyanide-bridged transition metal ions. See Vaucher et al. Angew. Chem. Int. Ed., 39, 1793 (2000); Vaucher et al., Nano Lett, 2, 225 (2002); Uemura and Kitagawa, J. Am. Chem. Soc.
  • Reverse microemulsions comprise nanometer scale water droplets stabilized in an organic phase by a surfactant, which can be anionic, cationic, or neutral in charge.
  • a surfactant which can be anionic, cationic, or neutral in charge.
  • W value i.e., [H 2 O]/[surfactant]
  • the presently disclosed subject matter provides a method of synthesizing a nanoparticle comprising a coordination polymer for use as an anticancer therapeutic agent or a multi-modal agent.
  • the presently disclosed synthesis methods involve the use of microemulsions in preparing NCPs.
  • the microemulsion can be water-in-oil (i.e., reverse micelles or water droplets dispersed in oil), oil-in-water (i.e., micelles or oil droplets dispersed in water), or a bi-continuous system containing comparable amounts of two immiscible fluids.
  • microemulsions can be made by mixing together two non-aqueous liquids of differing polarity with negligible mutual solubility.
  • the immiscible liquids that can be used to make the microemulsion typically include a relatively polar (i.e., hydrophobic) liquid and a relative non- polar (i.e., hydrophillic) liquid. While a large variety of polar/non-polar liquid mixtures can be used to form a microemulsion useful in the invention, the choice of particular liquids utilized can depend on the type of nanoparticles being made. Upon a review of the instant disclosure, a skilled artisan can select specific liquids for particular applications by adapting known methods of making microemulsions for use in the presently disclosed methods. In many embodiments, the relatively polar liquid is water, although other polar liquids might also be useful.
  • Non-polar liquids include alkanes (e.g., any liquid form of hexane, heptane, octane, nonane, decane, undecane, dodecane, etc.), cycloalkanes (e.g., cyclopentane, cyclohexane, etc.), aromatic hydrocarbons (e.g., benzene, toluene, etc.), and mixtures of the foregoing (e.g., petroleum and petroleum derivatives).
  • any such non-polar liquid can be used as long as it is compatible with the other components used to form the microemulsion and does not interfere with any precipitation reaction used to isolate the particles after their preparation.
  • surfactants are surface active agents that thermodynamically stabilize the very small dispersed micelles or reverse micelles in microemulsions.
  • surfactants possess an amphipathic structure that allows them to form films with very low interfacial tension between the oily and aqueous phases.
  • any substance that reduces surface tension at the interface of the relatively polar and relatively non-polar liquids and is compatible with other aspects of the presently disclosed subject matter can be used to form the microemulsion used to make nanoparticles.
  • the choice of a surfactant can depend on the particular liquids utilized and on the type of nanoparticles being made.
  • surfactants suitable for particular applications can be selected from known methods of making microemulsions or known characteristics of surfactants.
  • non-ionic surfactants are generally preferred when an ionic reactant is used in the microemulsion process and an ionic detergent would bind to or otherwise interfere with the ionic reactant.
  • soaps such as potassium oleate, sodium oleate, etc.
  • anionic detergents such as sodium cholate, sodium caprylate, etc.
  • cationic detergents such as cetylpyridynium chloride, alkyltrimethylammonium bromides, benzalkonium chloride, cetyldimethylethylammonium bromide, etc
  • zwitterionic detergents such as N-alkyl-N,N-dimethylammonio-1-propanesulfonates and CHAPS
  • non-ionic detergents such as polyoxyethylene esters, and various tritons (e.g., (TRITONTM-X100, TR!TONTM-X114); etc.
  • concentration of surfactant used can depend on many factors including the particular surfactant selected, liquids used, and the type of nanoparticles to be made. Suitable concentrations can be determined empirically; i.e., by trying different concentrations of surfactant until the concentration that performs best in a particular application is found. Ranges of suitable concentrations can also be determined from known critical micelle concentrations.
  • the presently disclosed subject matter provides a method of preparing a nanoscale coordination polymer comprising a plurality of platinum metal complexes where the method comprises: providing a first mixture comprising a microemulsion system comprising water, an organic solvent, a surfactant, and a co- surfactant; adding to the first mixture an aqueous solution comprising a platinum metal complex, thereby forming a second mixture, wherein the platinum metal complex comprises a platinum metal atom, one or more nonbridging ligands, and at least one ligand bound to the platinum metal atom by at least one coordination bond and comprising at least one prelinking moiety, wherein the at least one prelinking moiety is a group that can form a coordination bond with an additional metal atom; and stirring the second mixture for a period of time, thereby synthesizing the nanoparticle.
  • the presently disclosed subject matter provides a method of preparing a bimetallic nanoscale coordination polymer using microemulsion techniques.
  • the presently disclosed subject matter provides a method of synthesizing a nanoparticle comprising a coordination polymer comprising a plurality of platinum metal complexes, the method comprising: providing a first mixture comprising a microemulsion system comprising water, an organic solvent, a surfactant, and a co- surfactant; adding to the first mixture an aqueous solution comprising a platinum metal complex, thereby forming a second mixture, wherein the platinum metal complex comprises a platinum metal atom, one or more nonbridging ligands, and at least one ligand bound to the platinum metal atom by at least one coordination bond and comprising at least one prelinking moiety, wherein the at least one prelinking moiety is a group that can form a coordination bond with an additional metal atom; stirring the second mixture until the second mixture is visably clear; providing a
  • the nonplatinum metal compound can comprise a nonplatinum metal atom which can be a nonplatinum transition metal atom, a lanthanide metal atom, an actinide metal atom or combinations thereof.
  • the nonplatinum metal atom can be a paramagentic metal atom useful in MRI imaging.
  • the nonplatinum metal atom could also be a metal atom useful in a PET or SPECT imaging agent.
  • the nonplatinum metal atom is Tb 3+ OrZn 2+ .
  • the nonplatinum metal compound is TbCI 3 .
  • the mixing comprises stirring (e.g., using a magnetic stirrer or a mechanical stirrer). Mixing can also refer to sonication or to manual or mechanical shaking, or to any combination thereof.
  • the surfactant is a non-ionic surfactant. In some embodiments, the surfactant is TRITONTM -X100. In some embodiments, the co-surfactant is 1-hexanol. In some embodiments, the molar ratio of TRITONTM -X100 to 1-hexanol ranges between about 1 and about 5.
  • useful water to surfactant ratios i.e., I/I/, the ratio of [water]/[surfactant]
  • the third mixture i.e., afterthe addition of the polymerization agent, which can contribute to the water content of the mixture if dissolved in an aqueous carrier
  • varying I/I/ can lead to variations in the size of the resulting nanoparticles.
  • the presently disclosed subject matter also provides microemulsion methods of preparing nanoscale coordination polymers comprising polymeric bridging ligands.
  • the presently disclosed subject matter provides a method of synthesizing a nanoparticle comprising a coordination polymer comprising a plurality of platinum metal complexes, wherein the method comprises providing a microemulsion system comprising water; an organic solvent; a surfactant; a co-surfactant; a polymerizable monomer; and a platinum metal complex, wherein the platinum metal complex comprises a platinum metal atom, one or more nonbridging ligands, and at least one ligand bonded to the platinum metal atom by at least one coordination bond and comprising at least one prelinking moiety, where the at least one prelinking moiety is a moiety that can react with the polymerizable monomer.
  • a polymerization agent can be added to the microemulsion system to initiate polymerization of the polymerizable monomer.
  • Suitable prelinking moieties include, but are not limited to, alkyl halides, acyl halides, silyl ethers, alkenes, alkynes, carboxylic acids, amines, esters, anhydrides, and isocyanates.
  • Suitable polymerizable monomers include, but are not limited to, silyl ethers (e.g., TEOS), acrylic acid, and acrylamide.
  • TEOS is the polymerizable monomer
  • the polymerization agent can be aqueous ammonia.
  • Other suitable polymerization agents include aqueous hydroxide (e.g., NaOH) or hydrazine.
  • a suitable polymerization agent is tetramethylethane diamine (TMEDA).
  • a suitable surfactant is cetyltimethyl ammonium bromide (CTAB).
  • CTAB cetyltimethyl ammonium bromide
  • NCPs prepared by precipitation or microemulsion techniques can be isolated from the solutions used in their synthesis via any suitable technique (i.e., filtration, decanting, lyophilization, evaporation or vacuum evaporation of the solutions). In some embodiments, the isolation of the nanoparticles can be done by centrifugation.
  • the method of synthesizing the nanoparticle comprises further steps to graft additional components onto the surface of the nanoparticle or to coat the nanoparicle with an outer layer.
  • the methods can further comprise coating the nanoparticle with one or more of a metal oxide, a lipid bilayer, an organic polymer, a silica-based polymer, and combinations thereof.
  • Silica-based polymers can be coated onto the nanoparticles by sol-gel techniques known in the art.
  • the method of synthesizing the nanoparticle further comprises grafting onto the surface of the nanoparticle one or more of a photosensitizer, a radiosensitizer, a radionuclide, an imaging agent, a passivating agent, and a targeting agent.
  • Thermogravimetric analysis was performed using a Shimadzu TGA-50 (Shimadzu Corp., Kyoto, Japan) equipped with a platinum pan and heated at a rate of 3°C/min under air.
  • Powder X-ray diffraction (PXRD) patterns were collected on a Bruker SMARTAPEX Il diffractometer (Bruker AXS, Inc., Madison, Wisconsin, United States of America) using Cu radiation. The PXRD patterns were processed with the APEX 2 package using phase ID plugin.
  • TEM micrographs were obtained on carbon-coated copper grids.
  • a Beckman Coulter N5 Submicron Particle Size Analyzer (Beckman Coulter, Fullerton, California, United States of America) was used to determine the sample's hydrodynamic diameter and polydispersity.
  • a Varian 820-MS Inductively Coupled Plasma-Mass Spectrometer (ICP-MS; Varian, Inc., Palo Alto, California, United States of America) was used to determine metal concentration.
  • Example 1.2 Dichloro(1 R,2R-cyclohexanediammine)-platinum(ll), Pt(R 1 R- DACH)CI 2 .
  • Example 1.3 Synthesis of Diaqua(1 R,2R-cyclohexanediammine)platinum(ll), [Pt(R, R-DACH)(H 2 O) 2 ] 2+ .
  • Example 1.4 Preparation of Pt(II) NCPs via the Addition of an Initiator/Poor Solvent.
  • a precursor solution was prepared by mixing the platinum(ll) diaqua complex and the bridging ligand in distilled water to reach a final solute concentration on the order of 10.3 M. At this time the pH of the solution could be adjusted via the addition of dilute NaOH or HCI. A poor solvent was rapidly added to the precursor solution to initiate precipitation of the desired product. The nanoparticles were isolated via centrifugation and washed with ethanol before redispersement in ethanol via sonication.
  • the pH of a mixture of [cis Pt(NHs) 2 (OH 2 ⁇ ] 2+ (1 0 ⁇ mol) and benzene dicarboxylate (BDC, 1.0 ⁇ mol) in 200.0 ⁇ l_ distilled water was adjusted to 5.5 with dilute NaOH.
  • Acetone was rapidly added to the magnetically stirred precursor solution, which resulted in the formation of a clear solution that was bluish-white in color.
  • the resulting mixture was magnetically stirred in the dark for an additional 1 h before isolating the product via centrifugation, washing with ethanol, and redispersing in ethanol via sonication.
  • Figure 9 shows the SEM micrographs of c/s-Pt(NH 3 ) 2 (BDC) nanoclusters synthesized via the rapid addition of acetone to a precursor (aqueous) solution of the components.
  • Figure 10 shows the SEM micrographs of c/s-Pt(NH 3 ) 2 (BDC) nanoclusters synthesized via the rapid addition of an acetone:ethanol mixture (1 :1 v/v) to a precursor (aqueous) solution of the components.
  • Example 2.1.5 Synthesis of 2,2'-Bipyridine-5,5'-dicarboxylic acid.
  • a mixture of 5,5'-dimethyl- 2,2'-bipyridine and KMnO 4 in water (500 ml_) was heated at 70 °C for 24 h with magnetic stirring.
  • the brown precipitate was removed by filtration and washed multiple times with aqueous NaOH (1 M).
  • the combined water fractions were extracted with CHCI 3 to remove unreacted starting material and subsequently neutralized with aqueous HCI (2 M). After concentrating the solution to a volume of approximately 300 mL, the solution was acidified further to a pH of 6 to precipitate the product.
  • the light blue product was collected by centrifugation, washed with ethanol, and dried in vacuo.
  • Example 2.1.6 Synthesis of Dichloro(2,2'-bipyridine-5,5'- dicarboxylato)platinum(ll).
  • Example 2.2.1 Microemulsion Synthesis of Tb x [c,c,f- Pt(NH3)2CI 2 (O 2 CCH2CH2C ⁇ 2)2]y coordination polymers.
  • Nanometer-scale lanthanide-based coordination polymers with Pt(IV)- containing bridging complexes were prepared using a cationic cetyltrimethylammonium bromide (CTAB)/1-hexanol/iso-octane/H 2 O microemulsion system. Briefly, a round bottom flask was charged with CTAB and a particular volume of 0.50 M 1-hexanol / isooctane solution to yield a milky white mixture with a CTAB concentration of 0.050 M.
  • CTAB cetyltrimethylammonium bromide
  • Nanometer-scale coordination polymers with Pt(IV)-containing bridging complexes were prepared using a cationic cetyltrimethylammonium bromide (CTAB)/1-hexanol/iso-octane/H 2 O microemulsion system. Briefly, a round bottom flask was charged with CTAB and a particular volume of 0.50 M Ihexanol in isooctane solution to yield a milky white mixture with a CTAB concentration of 0.05 M.
  • CTAB cetyltrimethylammonium bromide
  • the resulting mixture was magnetically stirred in the dark for an additional 1 h before isolating the product via centrifugation, washing with methanol and ethanol, and redispersing in ethanol via sonication. Yield: 235 mg (73% isolated based on DSCP).
  • the NCPs exhibited a spherical morphology and were structurally amorphous, yielding no PXRD peaks that would indicate a crystalline phase.
  • the composition of the particles was deduced from ICP-MS measurements and TGA data. ICP-MS measurements gave an approximate Tb:Pt molar ratio of 2:3, which is expected for the charge balanced formula Tb 2 (DSCP) 3 (H 2 O) x .
  • the number of water molecules was determined from TGA data. There was an approximately 9.2% weight loss for the water species and an approximately 36.0 % weight loss for the organic species.
  • the sum MW of the organic species (i.e., 2 NH 3 and 2 succinate groups) for DSCP was determined to be approximately 268.
  • the approximate TGA formula weight of 1115 (268 x 1.5/.36) was determined.
  • Figure 12 shows the SEM micrographs of Tb x [C, c,t-
  • NCP-1 Pt(NH 3 ) 2 CI 2 (O 2 CCH 2 CH 2 CO 2 ) 2 ] y
  • NCP-1 Pt(NH 3 ) 2 CI 2 (O 2 CCH 2 CH 2 CO 2 ) 2 ] y
  • DLS measurements gave a diameter of 58.3 ⁇ 11.3 nm for the NCP-1 particles.
  • the NCPs were stable and readily dispersible in most organic solvents. NCP formation was reversible if excess water was added to the reaction mixture or to the isolated particles.
  • the Tb x [c,c>Pt(NH 3 ) 2 CI 2 (O 2 CCH 2 CH 2 CO 2 ) 2 ] y nanoparticles can be synthesized by rapid addition of a 1 :1 ethanol/methanol solution fo an aqueous solution of [c,c, f-Pt(NH 3 ) 2 CI 2 (succ) 2 ]-2 N(CH 3 )H 2 and TbCI 3 .
  • Figure 13 shows SEM images of these Tb x [c,c,f-Pt(NH 3 ) 2 CI 2 (O 2 CCH 2 CH 2 CO 2 ) 2 ] y nanoparticles.
  • a precursor solution was prepared by mixing the dimethylammonium salt of the platinum complex and the second metal in distilled water to reach a final solute concentration on the order of 10.3 M. At this time the pH of the solution could be adjusted via the addition of dilute NaOH or HCI. An initiator solvent was rapidly added to the precursor solution to initiate precipitation of the desired product. The nanoparticles were isolated via centrifugation and washed with ethanol before redispersement in ethanol via sonication.
  • Figure 14 shows the SEM micrographs of Zn x [C, c,t- Pt(NHs) 2 CI 2 (O 2 CCH 2 CH 2 CO 2 H nanoparticles synthesized via the rapid addition of methanol to a precursor (aqueous) solution of the components.
  • the pale yellow solid was suspended in approximately 2 ml_ of distilled H 2 O, and the pH of the suspension was adjusted to approximately 7 by titrating with dilute KOH.
  • the resulting solution was dialyzed against water using 3500 MW cutoff dialysis tubing.
  • Example 3.3 Assembly of Nanoparticles from Polymers Linked by Metal Ions.
  • a precursor solution of the polymers described in Examples 3.2.1 or 3.2.2 and metal ion was prepared, and nanoparticles were subsequently precipitated via the addition of poor (initiator) solvent or using microemulsion methods as previously described.
  • Example 4.1 Preparation of Polyvinylpyrolidone (PCP) Modified/Stabilized NCPs (NCP-I).
  • PCP Polyvinylpyrolidone
  • NCP-I PolyvinylpyroHidone
  • An ethanolic dispersion of NCP-1 was diluted to a final concentration of between about 2 and about 5 mg/mL with absolute ethanol.
  • PVP polyvinylpyroHidone
  • the PVP modified NCPs were isolated via centrifuge, washed with ethanol, and redispersed in ethanol.
  • Figure 15 shows the TEM micrographs of PVP-modified NCP-1 prepared according to the presently disclosed methods.
  • NCP-1 particles were coated with silica using sol-gel methodology. See Graf et al., Langmuir, 19, 6693-6700 (2003). An aliquot of the ethanolic dispersion of PVP-modified NCP-1 was diluted to a concentration of 0.2 mg/mL in 4% (v/v) NH 3 in ethanol. An aliquot of tetraethyl orthosilicate (TEOS, 2.5 ⁇ L/1.0 mg) was added to the reaction with magnetic stirring and the silica shell was allowed to evolve for at least 2 hours. Silica shell thickness typically increases with time and the volume of TEOS added to the reaction mixture.
  • TEOS tetraethyl orthosilicate
  • FIGS 14A-14D show the TEM micrographs of silica-coated NCP-1 isolated after 2 hours (i.e., NCP-1 '-a) ( Figure 16A), three hours ( Figure 16B), four hours (i.e., NCP-1 '-b) ( Figure 16C) and 7 hours ( Figure 16D) exposure to TEOS.
  • NCP-1 '-a particles had a silica layer thickness of about 2 nm (overall DLS diameter of 52.8 ⁇ 8.1 nm), while the NCP-1 '-b particles had a silica layer thickness of about 7 nm (68.6 ⁇ 10.2 nm).
  • Figure 18 shows the TGA curve for the DSCP molecular complex, NCP-1 nanoparticles, and NCP-1 ' particles. TGA gave a 7.0 and 8.5 % reduction in the total weight loss for NCP-1 '-a and NCP-1 '-b, confirming the presence of the silica shell. The shell thickness was highly reproducible, varying only to a slight degree when the same reaction conditions were used on different samples.
  • % released ⁇ [V to t x C) + Y]/Z ⁇ x 100
  • V tot total solvent volume remaining
  • C Pt concentration as determined by ICP-MS
  • Y total mole Pt removed from solution
  • Z Pt added to dialysis tubing.
  • NCP-1'-a and NCP-1'-b were determined to be about 5.5 and about 9 hours, respectively. See Figure 19. These rates should allow sufficient time for the Pt- based NCPs to circulate throughout the body and accumulate in tumor tissue. See Matsumura et al.. Cancer Res., 46, 6387 (1986).
  • Non-silica-coated NCPs i.e., NCP-1 gave a half-life of dissolution of about 0.1 h.
  • cyclic RGDfK (c(RGDfK); 2.0 mg, 3.313 ⁇ mol) was added to a small round-bottom flask and dried under high vaccum for 1 hour.
  • Anhydrous DMSO 500 ⁇ l_
  • triethylamine (0.20 ⁇ l_) were added to the round-bottom flask, followed by 0.86 ⁇ l_ (3.44 ⁇ mol) of (3- isocyanatopropyl)triethoxysilane.
  • the mixture was magnetically stirred under argon gas for 24 hours.
  • the solution (4 mg c(RGDfK)/mL DMSO) was placed in a freezer for later use.
  • silyl- derivative molecule e.g., the silyl derivative of a photosensitizer, radiosensitizer, passivating agent, imaging agent, or targeting agent
  • the desired weight % (up to 10% by mass) of the silyl- derivative molecule was added to the dispersion, and magnetic stirring was continued for an additional 20 h.
  • the functionalized NCPs were isolated via centrifuge, washed with ethanol and DMSO, and dispersed in DMSO via sonication.
  • Example 6 In Vitro Activity of NCPs Cell Lines All cell lines were purchased from the Tissue Culture Facility of the UNC Lineberger Comprehensive Cancer Center (Chapel Hill, North Carolina, United States of America) and cultured as per American Type Culture Collection (ATCC; Manassas, Virginia, United States of America) recommendations.
  • HT29 cells ATCC# HTB 38
  • McCoy's 5A Cellgro, Manassas, Virginia, United States of America
  • MCF-7 cells (ATCC# HTB 22) were propagated in MEM Alpha (Cellgro, Manassas, Virginia, United States of America) supplemented with 10% fetal bovine serum, 1% penicillin/streptomycin, 1 % sodium pyruvate, and 10 ⁇ g/mL insulin.
  • HT-29 viability assays Angiogenic human colon cancer (HT-29) cells were grown in 96 well plates at 2000 cells/well to 100 ⁇ l_ total volume. After a 24 hour incubation, the media was replaced with 100 ⁇ l_ of drug solution containing 0.5% DMSO and 0.5% phosphate buffered saline (PBS) in media, with drug concentrations varying as indicated. All concentrations were performed in quadruplicate. Cell viability was measured after 72 hours using CellTiter 96 Aqueous One Solution Assay (Promega Corporation, Madison, Wisconsin, United States of America), according to the manufacturer's protocol.
  • NCP-1 and NCP-1' did not lead to any appreciable cell death, presumably because the DSCP species released from NCP-1 and NCP-1 S do hot have a pathway to enter the cells effectively. Further, there were no reductants in the media under the in vitro conditions used to transform DSCP into the active Pt(II) species. The released DSCP would become active in vivo through reduction with endogenous biomolecules, such as glutathione.
  • MCF-7 Viability Assays were also performed using MCF-7 cells (breast cancer). MCF-7 cells were assayed under the same conditions as HT-29, and also at 4000 cells/well and 72 hours.
  • DSCP is roughly equivalent to cisplatin in activity at 4000 cells/well, with an IC 50 of 11.1 ⁇ M for DSCP versus 10.7 ⁇ M for cisplatin.
  • NCP-1'-b was also tested but at 2000 cells/well. It gave an IC 50 value of 1.8 ⁇ M versus 2.1 ⁇ M for cisplatin, as shown in Figure 21 B.
  • MCF-7 does not overexpress the ⁇ v ⁇ 3 integrin, and so c(RGDfK) targeting was not necessary.

Abstract

La présente invention concerne des polymères de coordination à l’échelle nanométrique destinés à être utilisés comme agents anticancéreux et comme agents à double action anticancéreuse et d’imagerie. Les polymères de coordination à l’échelle nanométrique peuvent comprendre une pluralité de complexes des métaux du platine, des ligands de pontage à des médicaments anticancéreux ne contenant pas de platine complexés à de multiples centres métalliques, ou des combinaisons de ceux-ci. Les polymères de coordination à l’échelle nanométrique peuvent être ciblés de façon à être administrés à des cellules cancéreuses. Ils peuvent également comprendre des agents de stabilisation pour permettre une libération contrôlée et/ou prolongées d’agents anticancéreux in vivo.
PCT/US2009/034867 2008-02-22 2009-02-23 Nanoparticules hybrides comme agents thérapeutiques anticancéreux et agents doubles thérapeutiques/de contraste d’imagerie WO2009139939A2 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/918,748 US20110135571A1 (en) 2008-02-22 2009-02-23 Hybrid nanoparticles as anti-cancer therapeutic agents and dual therapeutic/imaging contrast agents

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US3074608P 2008-02-22 2008-02-22
US61/030,746 2008-02-22
US13756508P 2008-07-31 2008-07-31
US61/137,565 2008-07-31

Publications (2)

Publication Number Publication Date
WO2009139939A2 true WO2009139939A2 (fr) 2009-11-19
WO2009139939A3 WO2009139939A3 (fr) 2010-03-18

Family

ID=41319229

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/034867 WO2009139939A2 (fr) 2008-02-22 2009-02-23 Nanoparticules hybrides comme agents thérapeutiques anticancéreux et agents doubles thérapeutiques/de contraste d’imagerie

Country Status (2)

Country Link
US (1) US20110135571A1 (fr)
WO (1) WO2009139939A2 (fr)

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101787053A (zh) * 2010-03-05 2010-07-28 复旦大学 铂配合物及其制备方法和药用用途
WO2011133925A2 (fr) * 2010-04-23 2011-10-27 Massachusetts Institute Of Technology Administration cytosolique photoactivée et ciblée de médicament
CN102584900A (zh) * 2012-01-20 2012-07-18 沈阳化工大学 贵金属配位化合物及其制备方法
FR2971942A1 (fr) * 2011-02-28 2012-08-31 Centre Nat Rech Scient Systeme generant des especes reactives de l'oxygene pour utilisation comme medicament dans le traitement du cancer
EP2371926A3 (fr) * 2010-03-22 2012-12-05 Samsung Display Co., Ltd. Dispositif électroluminescent incluant un hybride de copolymère en bloc à points quantiques et son procédé de fabrication
WO2013009701A2 (fr) 2011-07-08 2013-01-17 The University Of North Carolina At Chapel Hill Nanoparticules de métal-bisphosphonate pour thérapie anticancéreuse et imagerie, ainsi que pour traiter des troubles des os
WO2013112823A1 (fr) * 2012-01-25 2013-08-01 Dow Corning Corporation Synthèse d'hétérophase de matériaux à structure métal - coordonné pour la régulation de taille de particule
CN105214089A (zh) * 2014-06-03 2016-01-06 复旦大学附属华山医院 一种具有核壳结构的磁性胶体纳米晶簇及其制备方法和应用
CN108559100A (zh) * 2018-05-16 2018-09-21 大连理工大学 金属离子引导羧酸配体官能化多酸化合物及其制备方法与催化降解化学战剂模拟物的应用
US10206871B2 (en) 2014-10-14 2019-02-19 The University Of Chicago Nanoparticles for photodynamic therapy, X-ray induced photodynamic therapy, radiotherapy, chemotherapy, immunotherapy, and any combination thereof
CN110384703A (zh) * 2019-07-19 2019-10-29 湖南大学 基于6-硫鸟嘌呤的金属-药物配位纳米药物及其制备方法和应用
WO2019218275A1 (fr) * 2018-05-16 2019-11-21 大连理工大学 Composé de polyacide guidé par des ions métalliques et fonctionnalisé par un ligand carboxylate, procédé de préparation s'y rapportant et son utilisation dans la dégradation catalytique de simulateurs d'agents de guerre chimique
US10517822B2 (en) 2013-11-06 2019-12-31 The University Of Chicago Nanoscale carriers for the delivery or co-delivery of chemotherapeutics, nucleic acids and photosensitizers
CN111194232A (zh) * 2017-08-02 2020-05-22 芝加哥大学 用于x射线诱导的光动力学疗法、放射疗法、放射动力学疗法、化学疗法、免疫疗法以及它们的任何组合的纳米级金属有机层和金属有机纳米片
US10806694B2 (en) 2014-10-14 2020-10-20 The University Of Chicago Nanoparticles for photodynamic therapy, X-ray induced photodynamic therapy, radiotherapy, radiodynamic therapy, chemotherapy, immunotherapy, and any combination thereof
CN112870355A (zh) * 2021-01-28 2021-06-01 中山大学 一种复合纳米多孔的铂基配位聚合物及其制备方法和应用
US20220033266A1 (en) * 2019-02-28 2022-02-03 Anhui University Copper ion-doped carbon dots, preparation method and application thereof as photosensitizer for photodynamic therapy
US11246877B2 (en) 2016-05-20 2022-02-15 The University Of Chicago Nanoparticles for chemotherapy, targeted therapy, photodynamic therapy, immunotherapy, and any combination thereof

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8945527B2 (en) * 2008-04-25 2015-02-03 The University Of North Carolina At Chapel Hill Degradable compounds and methods of use thereof, particularly with particle replication in non-wetting templates
US8309489B2 (en) * 2009-06-18 2012-11-13 University Of Central Florida Research Foundation, Inc. Thermally stable nanoparticles on supports
US8992984B1 (en) 2009-10-21 2015-03-31 Stc.Unm Protocells and their use for targeted delivery of multicomponent cargos to cancer cells
WO2012037358A1 (fr) 2010-09-16 2012-03-22 The University Of North Carolina At Chapel Hill Monomères de silyle bifonctionnel asymétrique et leurs particules, utilisés comme promédicaments et excipients d'administration d'agents pharmaceutiques, chimiques et biologiques
US8952119B2 (en) 2010-11-18 2015-02-10 Aspen Aerogels, Inc. Organically modified hybrid aerogels
WO2013082304A1 (fr) * 2011-11-29 2013-06-06 The Regents Of The University Of California Reconfiguration de circuit génique à médiation par une nanoantenne photonique
US20140205543A1 (en) * 2013-01-24 2014-07-24 Memorial Sloan-Kettering Cancer Center Method for diagnosing or treating tumors using sphingomyelin containing liposomes
JP6479799B2 (ja) * 2013-08-13 2019-03-06 アンテオ テクノロジーズ プロプライエタリー リミテッドAnteo Technologies Pty Ltd 粒子への分子の接合
US11672866B2 (en) 2016-01-08 2023-06-13 Paul N. DURFEE Osteotropic nanoparticles for prevention or treatment of bone metastases
US11344629B2 (en) 2017-03-01 2022-05-31 Charles Jeffrey Brinker Active targeting of cells by monosized protocells
US20190283137A1 (en) * 2018-03-14 2019-09-19 Lawrence Livermore National Security, Llc Metallopolymers for additive manufacturing of metal foams
US20190314324A1 (en) 2018-04-13 2019-10-17 The University Of Chicago Combination of micheliolide derivatives or nanoparticles with ionizing radiation and checkpoint inhibitors for cancer therapy
TWI808178B (zh) * 2019-05-21 2023-07-11 隆達電子股份有限公司 波長轉換物質及發光裝置
CN110862546B (zh) * 2019-10-12 2021-07-09 厦门大学 一种甲氨蝶呤金属配位聚合物及其制备方法和应用
CN112442117B (zh) * 2020-12-14 2022-07-08 复旦大学 一种靶向卵泡刺激素受体的肿瘤成像与治疗探针及其制备方法与应用
CN113588523B (zh) * 2021-07-26 2022-03-29 浙江大学 一种用于质谱流式细胞技术的基于框架结构的纳米颗粒及其制备方法
CN114939165B (zh) * 2022-05-23 2023-05-30 河北工业大学 可逆转多药耐药性的双金属纳米粒及其制备方法和应用

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4405771A (en) * 1980-10-27 1983-09-20 Yeda Research & Development Co., Ltd. Organometallic polymers, their preparation and compositions containing them
WO2007090295A1 (fr) * 2006-02-09 2007-08-16 Simon Fraser University Polymeres de coordination birefringents contenant des metaux
WO2007108618A1 (fr) * 2006-03-17 2007-09-27 Soon Hai Hong Nanoparticules organométalliques hydrosolubles et leur procédé de préparation

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5147806A (en) * 1988-04-29 1992-09-15 Igen, Inc. Method and apparatus for conducting electrochemiluminescence measurements
US5213788A (en) * 1988-09-29 1993-05-25 Ranney David F Physically and chemically stabilized polyatomic clusters for magnetic resonance image and spectral enhancement
JP2611073B2 (ja) * 1990-07-26 1997-05-21 モンサント カンパニー 新規ポリアミンの製造方法
US6013638A (en) * 1991-10-02 2000-01-11 The United States Of America As Represented By The Department Of Health And Human Services Adenovirus comprising deletions on the E1A, E1B and E3 regions for transfer of genes to the lung
US5858784A (en) * 1991-12-17 1999-01-12 The Regents Of The University Of California Expression of cloned genes in the lung by aerosol- and liposome-based delivery
US5871710A (en) * 1992-09-04 1999-02-16 The General Hospital Corporation Graft co-polymer adducts of platinum (II) compounds
US5641623A (en) * 1995-01-04 1997-06-24 Martin; Mark T. Electrochemiluminescence assay
US20010003647A1 (en) * 1995-06-07 2001-06-14 Ji Sun Coreatant-including electrochemiluminescent compounds, methods, systems and kits utilizing same
US6022737A (en) * 1995-11-02 2000-02-08 Amgen Inc. Formulations for non-viral in vivo transfection in the lungs
US5874064A (en) * 1996-05-24 1999-02-23 Massachusetts Institute Of Technology Aerodynamically light particles for pulmonary drug delivery
US6180082B1 (en) * 1997-11-24 2001-01-30 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Method to enhance tissue accumulation of radiolabeled compounds
US6602932B2 (en) * 1999-12-15 2003-08-05 North Carolina State University Nanoparticle composites and nanocapsules for guest encapsulation and methods for synthesizing same
US6530944B2 (en) * 2000-02-08 2003-03-11 Rice University Optically-active nanoparticles for use in therapeutic and diagnostic methods
US6548264B1 (en) * 2000-05-17 2003-04-15 University Of Florida Coated nanoparticles
KR100401335B1 (en) * 2003-03-08 2003-10-10 Mijitech Co Ltd Metal nanoparticle surface-coated with silicon oxides and preparation thereof
WO2004084871A1 (fr) * 2003-03-26 2004-10-07 Ltt Bio-Pharma Co., Ltd. Nanoparticules intraveineuses pour cibler une administration de medicaments et pour une liberation de medicaments prolongee
WO2005011629A1 (fr) * 2003-07-31 2005-02-10 3M Innovative Properties Company Compositions bioactives comprenant des triazines
US20050147963A1 (en) * 2003-12-29 2005-07-07 Intel Corporation Composite organic-inorganic nanoparticles and methods for use thereof
CN1305937C (zh) * 2005-02-25 2007-03-21 复旦大学 一种金属配位聚合物纳米结构材料的制备方法
US20080124281A1 (en) * 2006-11-29 2008-05-29 Board Of Regents, The University Of Texas System Nanotubular probes as ultrasensitive mr contrast agent

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4405771A (en) * 1980-10-27 1983-09-20 Yeda Research & Development Co., Ltd. Organometallic polymers, their preparation and compositions containing them
WO2007090295A1 (fr) * 2006-02-09 2007-08-16 Simon Fraser University Polymeres de coordination birefringents contenant des metaux
WO2007108618A1 (fr) * 2006-03-17 2007-09-27 Soon Hai Hong Nanoparticules organométalliques hydrosolubles et leur procédé de préparation

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
DATABASE WPI Week 200629 Thomson Scientific, London, GB; AN 2006-273397 XP002564689 & CN 1 673 258 A (UNIV FUDAN) 28 September 2005 (2005-09-28) *
G.V. KALAYDA, E.A.: "Synthesis, structure and biological activity of new azine-bridged dinuclear platinum(ii) complexes" EUR. J. INORG. CHEM., 2003, pages 4347-4355, XP002564740 *
JOHN E SHEATS: "History of organometallic polymers" JOURNAL OF MACROMOLECULAR SCIENCE : PART A - CHEMISTRY, MARCEL DEKKER, NEW YORK, NY, US, vol. 15, no. 6, 1 January 1981 (1981-01-01), pages 1173-1199, XP008117421 ISSN: 0022-233X *
LEIGH G J: "Comprehensive coordination chemistry II From Biology to Nanotechnology" JOURNAL OF ORGANOMETALLIC CHEMISTRY, ELSEVIER-SEQUOIA S.A. LAUSANNE, CH, vol. 689, no. 16, 15 August 2004 (2004-08-15), pages 2733-2742, XP004523077 ISSN: 0022-328X *
WEIYONG YU, E.A.: "Immobilization of polymer-stabilized metal colloids by a modified coordination capture" JOURNAL OF MOLECULAR CATALYSIS A: CHEMICAL, vol. 142, 1999, pages 201-211, XP002564739 *

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101787053A (zh) * 2010-03-05 2010-07-28 复旦大学 铂配合物及其制备方法和药用用途
US8766315B2 (en) 2010-03-22 2014-07-01 Samsung Display Co., Ltd. Quantum dot-block copolymer hybrid, methods of fabricating and dispersing the same, light emitting device including the same, and fabrication method thereof
EP2371926A3 (fr) * 2010-03-22 2012-12-05 Samsung Display Co., Ltd. Dispositif électroluminescent incluant un hybride de copolymère en bloc à points quantiques et son procédé de fabrication
WO2011133925A3 (fr) * 2010-04-23 2012-03-22 Massachusetts Institute Of Technology Administration cytosolique photoactivée et ciblée de médicament
WO2011133925A2 (fr) * 2010-04-23 2011-10-27 Massachusetts Institute Of Technology Administration cytosolique photoactivée et ciblée de médicament
FR2971942A1 (fr) * 2011-02-28 2012-08-31 Centre Nat Rech Scient Systeme generant des especes reactives de l'oxygene pour utilisation comme medicament dans le traitement du cancer
EP2729180A4 (fr) * 2011-07-08 2015-03-04 Univ North Carolina Nanoparticules de métal-bisphosphonate pour thérapie anticancéreuse et imagerie, ainsi que pour traiter des troubles des os
US10596116B2 (en) 2011-07-08 2020-03-24 The University Of North Carolina At Chapel Hill Metal bisphosphonate nanoparticles for anti-cancer therapy and imaging and for treating bone disorders
WO2013009701A2 (fr) 2011-07-08 2013-01-17 The University Of North Carolina At Chapel Hill Nanoparticules de métal-bisphosphonate pour thérapie anticancéreuse et imagerie, ainsi que pour traiter des troubles des os
US11872311B2 (en) 2011-07-08 2024-01-16 The University Of North Carolina At Chapel Hill Metal bisphosphonate nanoparticles for anti-cancer therapy and imaging and for treating bone disorders
US9693957B2 (en) 2011-07-08 2017-07-04 The University Of North Carolina At Chapel Hill Metal bisphosphonate nanoparticles for anti-cancer therapy and imaging and for treating bone disorders
EP3494974A1 (fr) * 2011-07-08 2019-06-12 The University of North Carolina at Chapel Hill Nanoparticules de métal-bisphosphonate pour thérapie anticancéreuse et imagerie, ainsi que pour traiter des troubles des os
CN102584900A (zh) * 2012-01-20 2012-07-18 沈阳化工大学 贵金属配位化合物及其制备方法
WO2013112823A1 (fr) * 2012-01-25 2013-08-01 Dow Corning Corporation Synthèse d'hétérophase de matériaux à structure métal - coordonné pour la régulation de taille de particule
US10517822B2 (en) 2013-11-06 2019-12-31 The University Of Chicago Nanoscale carriers for the delivery or co-delivery of chemotherapeutics, nucleic acids and photosensitizers
CN105214089B (zh) * 2014-06-03 2018-08-28 复旦大学附属华山医院 一种具有核壳结构的磁性胶体纳米晶簇及其制备方法和应用
CN105214089A (zh) * 2014-06-03 2016-01-06 复旦大学附属华山医院 一种具有核壳结构的磁性胶体纳米晶簇及其制备方法和应用
US10206871B2 (en) 2014-10-14 2019-02-19 The University Of Chicago Nanoparticles for photodynamic therapy, X-ray induced photodynamic therapy, radiotherapy, chemotherapy, immunotherapy, and any combination thereof
US10780045B2 (en) 2014-10-14 2020-09-22 The University Of Chicago Nanoparticles for photodynamic therapy, X-ray induced photodynamic therapy, radiotherapy, chemotherapy, immunotherapy, and any combination thereof
US10806694B2 (en) 2014-10-14 2020-10-20 The University Of Chicago Nanoparticles for photodynamic therapy, X-ray induced photodynamic therapy, radiotherapy, radiodynamic therapy, chemotherapy, immunotherapy, and any combination thereof
US11246877B2 (en) 2016-05-20 2022-02-15 The University Of Chicago Nanoparticles for chemotherapy, targeted therapy, photodynamic therapy, immunotherapy, and any combination thereof
US11826426B2 (en) 2017-08-02 2023-11-28 The University Of Chicago Nanoscale metal-organic layers and metal-organic nanoplates for x-ray induced photodynamic therapy, radiotherapy, radiodynamic therapy, chemotherapy, immunotherapy, and any combination thereof
CN111194232B (zh) * 2017-08-02 2023-01-31 芝加哥大学 纳米级金属有机层和金属有机纳米片
CN111194232A (zh) * 2017-08-02 2020-05-22 芝加哥大学 用于x射线诱导的光动力学疗法、放射疗法、放射动力学疗法、化学疗法、免疫疗法以及它们的任何组合的纳米级金属有机层和金属有机纳米片
WO2019218275A1 (fr) * 2018-05-16 2019-11-21 大连理工大学 Composé de polyacide guidé par des ions métalliques et fonctionnalisé par un ligand carboxylate, procédé de préparation s'y rapportant et son utilisation dans la dégradation catalytique de simulateurs d'agents de guerre chimique
CN108559100B (zh) * 2018-05-16 2021-02-19 大连理工大学 金属离子引导羧酸配体官能化多酸化合物及其制备方法与催化降解化学战剂模拟物的应用
US11358133B2 (en) 2018-05-16 2022-06-14 Dalian University Of Technology Metal ion-directed carboxylic acid functionalized polyoxometalate hybrid compounds and their preparation method and applications for catalyzing the degradation of chemical warfare agent simulants
CN108559100A (zh) * 2018-05-16 2018-09-21 大连理工大学 金属离子引导羧酸配体官能化多酸化合物及其制备方法与催化降解化学战剂模拟物的应用
US20220033266A1 (en) * 2019-02-28 2022-02-03 Anhui University Copper ion-doped carbon dots, preparation method and application thereof as photosensitizer for photodynamic therapy
CN110384703A (zh) * 2019-07-19 2019-10-29 湖南大学 基于6-硫鸟嘌呤的金属-药物配位纳米药物及其制备方法和应用
CN112870355B (zh) * 2021-01-28 2022-04-01 中山大学 一种复合纳米多孔的铂基配位聚合物及其制备方法和应用
CN112870355A (zh) * 2021-01-28 2021-06-01 中山大学 一种复合纳米多孔的铂基配位聚合物及其制备方法和应用

Also Published As

Publication number Publication date
US20110135571A1 (en) 2011-06-09
WO2009139939A3 (fr) 2010-03-18

Similar Documents

Publication Publication Date Title
US20110135571A1 (en) Hybrid nanoparticles as anti-cancer therapeutic agents and dual therapeutic/imaging contrast agents
US11872311B2 (en) Metal bisphosphonate nanoparticles for anti-cancer therapy and imaging and for treating bone disorders
Luo et al. Metal–organic framework (MOF)-based nanomaterials for biomedical applications
Ovais et al. Designing stimuli‐responsive upconversion nanoparticles that exploit the tumor microenvironment
CN107001031B (zh) 一种金属有机框架、药物制剂及其在制备药物中的用途
US20090317335A1 (en) Hybrid Nanomaterials as Multimodal Imaging Contrast Agents
Fan et al. Intranuclear biophotonics by smart design of nuclear-targeting photo-/radio-sensitizers co-loaded upconversion nanoparticles
Zhang et al. Glutathione-capped fluorescent gold nanoclusters for dual-modal fluorescence/X-ray computed tomography imaging
Yang et al. Rodlike MSN@ Au nanohybrid-modified supermolecular photosensitizer for NIRF/MSOT/CT/MR quadmodal imaging-guided photothermal/photodynamic cancer therapy
Suarez-Garcia et al. Coordination polymers nanoparticles for bioimaging
Selvan Silica-coated quantum dots and magnetic nanoparticles for bioimaging applications (Mini-Review)
US20110238001A1 (en) Nanoparticle based photodynamic therapy and methods of making and using same
Rampazzo et al. NIR-fluorescent dye doped silica nanoparticles for in vivo imaging, sensing and theranostic
Li et al. Bright, magnetic NIR-II quantum dot probe for sensitive dual-modality imaging and intensive combination therapy of cancer
Zhang et al. pH-driven targeting nanoprobe with dual-responsive drug release for persistent luminescence imaging and chemotherapy of tumor
Yang et al. Gadolinium-based bimodal probes to enhance T1-Weighted magnetic resonance/optical imaging
Le et al. Emerging NIR light-responsive delivery systems based on lanthanide-doped upconverting nanoparticles
Yang et al. Core-shell structured nanoparticles for photodynamic therapy-based cancer treatment and related imaging
Addisu et al. Mixed lanthanide oxide nanoparticles coated with alginate-polydopamine as multifunctional nanovehicles for dual modality: Targeted imaging and chemotherapy
Xu et al. Advances in indocyanine green-based codelivery nanoplatforms for combinatorial therapy
Viana et al. 177Lu-Labeled Eu-doped mesoporous SiO2 nanoparticles as a theranostic radiopharmaceutical for colorectal cancer
Yu et al. Near-infrared light responsive upconversion nanoparticles for imaging, drug delivery and therapy of cancers
Wu et al. Quantum dots for cancer therapy and bioimaging
Icten Functional nanocomposites: promising candidates for cancer diagnosis and treatment
Steinborn et al. Metal-organic nanopharmaceuticals

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09747025

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 12918748

Country of ref document: US

122 Ep: pct application non-entry in european phase

Ref document number: 09747025

Country of ref document: EP

Kind code of ref document: A2